This invention relates to compounds based on the 3-deazapurines ring system and to methods for making such compounds. The invention also generally relates to the field of "antisense" agents, agents that are capable of specific hybridization with a nucleotide sequence of an RNA. The 3-deazapurine-containing compounds of this invention can be useful for modulating the activity of RNA when incorporated into oligonucleotides. Oligonucleotides and their analogs are used for a variety of therapeutic and diagnostic purposes, such as treating diseases, regulating gene expression in experimental systems, assaying for RNA and for RNA products through the employment of antisense interactions with such RNA, diagnosing diseases, modulating the production of proteins and cleaving RNA in site specific fashions.

Patent
   5750692
Priority
Mar 05 1993
Filed
Dec 10 1996
Issued
May 12 1998
Expiry
Mar 05 2013
Assg.orig
Entity
Small
327
6
EXPIRED
1. A method for preparing a 3-deazapurine nucleoside comprising the steps of:
a. alkylating a cyano-1-(3,5-di-O-p-toluoyl-β-D-pentose) imidazole in the presence of a base, said alkylating agent having the formula RX, wherein R is alkyl having up to about 12 carbon atoms, alkenyl having up to about 12 carbon atoms, or aralkyl having from about 6 to about 30 carbon atoms; and X is a leaving group;
b. ammonolyating the alkylated cyanoimidazole; and
c. cyclizing the ammonylated cyanoimidazole through basic catalysis at a temperature of greater than 90°C
4. A method for preparing a 3-deazapurine comprising the steps of:
a. providing an alkyl cyanoimidazole carboxylate;
b. alkylating the alkyl cyanoimidazole carboxylate alpha to the cyano group in the presence of a base, said alkylating agent having the formula RX, wherein R is alkyl having up to about 12 carbon atoms, alkenyl having up to about 12 carbon atoms, or arylalkyl having from about 6 to about 30 carbon atoms; and X is a leaving group;
c. ammonolyating the alkylated cyanoimidazole carboxylate; and
d. cyclizing the ammonolyated cyanoimidazole carboxamide through basic catalysis at a temperature of about 75°C
2. The method of claim 1 wherein the leaving group is a halogen.
3. The method of claim 1 wherein the cyclization is performed at about 95°C
5. The method of claim 4 wherein the leaving group is a halogen.
6. The method of claim 4 further comprising the step of protecting the cyanoimidazole carboxylate with a protecting group therefore.
7. The method of claim 6 wherein said protecting group is tetrahydropyranyl.
8. The method of claim 7 further comprising the step of acidifying the reaction mixture containing the cyanoimidazole carboxylate to remove the protecting groups.

This application is a divisional of U.S. patent application Ser. No. 08/514,762 filed Aug. 14, 1995, now U.S. Pat. No. 5,587,470, which is a divisional of U.S. patent application Ser. No. 08/027,011 filed Mar. 5, 1993, now U.S. Pat. No. 5,457,191, the disclosures of which are hereby incorporated in their entirety.

This invention relates to compounds that have utility as oligonucleotide intermediates, and to methods for making such compounds. The compounds are based on the 3-deazapurine core. The invention also generally relates to the field of "antisense" agents, agents that are capable of specific hybridization with a nucleotide sequence of an RNA. In particular, this invention relates to novel compounds that may be incorporated into oligonucleotides, these compounds include novel heterocyclic bases, nucleosides, and nucleotides. When incorporated into oligonucleotides, the 3-deazapurines of the invention can be useful for modulating the activity of RNA. Oligonucleotides are used for a variety of therapeutic and diagnostic purposes, such as treating diseases, regulating gene expression in experimental systems, assaying for RNA and for RNA products through the employment of antisense interactions with such RNA, diagnosing diseases, modulating the production of proteins, and cleaving RNA in site specific fashions.

It is well known that most of the bodily states in mammals including most disease states, are effected by proteins. Such proteins, either acting directly or through their enzymatic functions, contribute in major proportion to many diseases in animals and man. Classical therapeutics has generally focused upon interactions with such proteins in efforts to moderate their disease causing or disease potentiating functions. Recently, however, attempts have been made to moderate the actual production of such proteins by interactions with molecules that direct their synthesis, intracellular RNA. By interfering with the production of proteins, it has been hoped to effect therapeutic results with maximum effect and minimal side effects. It is the general object of such therapeutic approaches to interfere with or otherwise modulate gene expression leading to undesired protein formation.

One method for inhibiting specific gene expression is the use of oligonucleotides and oligonucleotide analogs as "antisense" agents. The oligonucleotides or oligonucleotide analogs complimentary to a specific, target, messenger RNA (mRNA) sequence are used. A number of workers have reported such attempts. Pertinent reviews include Stein, et al., Cancer Research, 1988, 48, 2659-2668; Walder, Genes & Development, 1988, 2, 502-504; Marcus-Sekura, Anal. Biochemistry, 1988, 172, 289-295; Zon, J. Protein Chemistry, 1987, 6, 131-145; Zon, Pharmaceutical Res., 1988, 5, 539-549; Van der Krol, et al., BioTechniques, 1988, 6, 958-973; and Loose-Mitchell, TIPS, 1988, 9, 45-47. Each of the foregoing provide background concerning general antisense theory and prior techniques.

Thus, antisense methodology has been directed to the complementary hybridization of relatively short oligonucleotides and oligonucleotide analogs to single-stranded mRNA or single-stranded DNA such that the normal, essential functions of these intracellular nucleic acids are disrupted. Hybridization is the sequence specific hydrogen bonding of oligonucleotides or oligonucleotide analogs to Watson-Crick base pairs of RNA or single-stranded DNA. Such base pairs are said to be complementary to one another.

Prior attempts at antisense therapy have provided oligonucleotides or oligonucleotide analogs that are designed to bind in a specific fashion to--which are specifically hybridizable with--a specific mRNA by hybridization. Such oligonucleotide and oligonucleotide analogs are intended to inhibit the activity of the selected mRNA--to interfere with translation reactions by which proteins coded by the MRNA are produced--by any of a number of mechanisms. The inhibition of the formation of the specific proteins that are coded for by the mRNA sequences interfered with have been hoped to lead to therapeutic benefits.

A number of chemical modifications have been introduced into antisense agents--oligonucleotides and oligonucleotide analogs--to increase their therapeutic activity. Such modifications are designed to increase cell penetration of the antisense agents, to stabilize the antisense agents from nucleases and other enzymes that degrade or interfere with their structure or activity in the body, to enhance the antisense agents' binding to targeted RNA, to provide a mode of disruption (terminating event) once the antisense agents are sequence-specifically bound to targeted RNA, and to improve the antisense agents' pharmacokinetic and pharmacodynamic properties. These modifications are designed to enhance the uptake of antisense agents--oligonucleotides and oligonucleotide analogs--and thus provide effective therapeutic, research reagent, or diagnostic uses.

Initially, only two mechanisms or terminating events have been thought to be operating in the antisense approach to therapeutics. These are the hybridization arrest mechanism and the cleavage of hybridized RNA by the cellular enzyme, ribonuclease H (RNase H). It is likely that additional "natural" events may be involved in the disruption of targeted RNA, however. These naturally occurring events are discussed by Cohen in oligonucleotides: Antisense Inhibitors of Gene Expression, CRC Press, Inc., Boca Raton, Fla. (1989).

The first, hybridization arrest, denotes the terminating event in which the oligonucleotide inhibitor binds to the target nucleic acid and thus prevents, by simple steric hindrance, the binding of essential proteins, most often ribosomes, to the nucleic acid. Methyl phosphonate oligonucleotides, Miller, et al., Anti-Cancer Drug Design, 1987, 2, 117-128, and α-anomer oligonucleotides are the two most extensively studied antisense agents that are thought to disrupt nucleic acid function by hybridization arrest.

The second "natural" type of terminating event is the activation of RNase H by the heteroduplex formed between the DNA type oligonucleotide or oligonucleotide analog and the targeted RNA with subsequent cleavage of target RNA by the enzyme. The oligonucleotides or oligonucleotide analogs, which must be of the deoxyribose type, hybridize with the targeted RNA and this duplex activates the RNase H enzyme to cleave the RNA strand, thus destroying the normal function of the RNA. Phosphorothioate modified oligonucleotides are the most prominent example of antisense agents that are thought to operate by this type of antisense terminating event. Walder, et al., in Proceedings of the National Academy of Sciences of the U.S.A., 1988, 85, 5011-5015 and Stein, et al., in Nucleic Acids Research, 1988, 16, 3209-3221 describe the role that RNase H plays in the antisense approach.

To increase the potency via the "natural" termination events the most often used oligonucleotide modification is modification at the phosphorus atoms. An example of such modifications include methyl phosphonate oligonucleotides, where the phosphoryl oxygen of the phosphorodiester linking moiety is replaced or the nucleotide elements together are replaced, either in total or in part, by methyl groups. Other types of modifications to the phosphorus atom of the phosphate backbone of oligonucleotides include phosphorothioate oligonucleotides. The phosphorothioate modified oligonucleotides are thought to terminate RNA by activation of RNase H upon hybridization to RNA although hybridization arrest of RNA function may play some part in their activity. Backbone modifications are disclosed as set forth in U.S. patent applications assigned to a common assignee hereof, entitled "Backbone Modified Oligonucleotide Analogs," Ser. No. 703,619 and "Heteroatomic Oligonucleotide Linkages," Ser. No. 903,160, the disclosures of which are incorporated herein by reference to disclose more fully such modifications. Phosphoroamidites have been disclosed as set forth in an application having U.S. application Ser. No. 918,362 and assigned to a common assignee hereof, entitled "Improved Process for Preparation of 2'-O-Alkylguanosines and Related Compounds," the disclosures of which are incorporated herein by reference to disclose more fully such modifications.

All applications of oligonucleotides and oligonucleotide analogs as antisense agents for therapeutic purposes, diagnostic purposes, and research reagents require that the oligonucleotides or oligonucleotide analogs be synthesized in large quantities, be transported across cell membranes or taken up by cells, appropriately hybridize to targeted RNA or DNA, and subsequently terminate or disrupt nucleic acid function. These critical functions depend on the initial stability of oligonucleotides and oligonucleotide analogs toward nuclease degradation.

A serious deficiency of unmodified oligonucleotides for these purposes, particularly antisense therapeutics, is the enzymatic degradation of the administered oligonucleotides by a variety of intracellular and extracellular ubiquitous nucleolytic enzymes, hereinafter referred to as "nucleases." It is unlikely that unmodified, "wild type," oligonucleotides will be useful therapeutic agents because they are rapidly degraded by nucleases. A primary focus of antisense research has been to modify oligonucleotides to render them resistant to such nucleases. These modifications have heretofore exclusively taken place on the sugar-phosphate backbone, particularly on the phosphorus atom. Phosphorothioates, methyl phosphonates, phosphoramidites, and phosphorotriesters (phosphate methylated DNA) have been reported to have various levels of resistance to nucleases. Backbone modifications are disclosed as set forth in U.S. patent applications assigned to a common assignee hereof, entitled "Backbone Modified Oligonucleotide Analogs," Ser. No. 703,619 and "Heteroatomic Oligonucleotide Linkages," Ser. No. 903,160, the disclosures of which are incorporated herein by reference to disclose more fully such modifications.

Other modifications to "wild type" oligonucleotides made to enhance resistance to nucleases, activate the RNase terminating event, and enhance the RNA-oligonucleotide duplex's hybridization properties include functionalizing the nucleoside's naturally occurring sugar. Sugar modifications are disclosed as set forth in PCT Application assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA Activity and Gene Expression," PCT Patent Application Number PCT\US91\00243, International Publication Number WO 91/10671, the disclosures of which are incorporated herein by reference to disclose more fully such modifications.

Other synthetic terminating events, as compared to hybridization arrest and RNase H cleavage, have been studied in an attempt to increase the potency of oligonucleotides and oligonucleotide analogs for use in antisense diagnostics and therapeutics. Thus, an area of research has developed in which a second domain to the oligonucleotide, generally referred to as a pendant group, has been introduced.

The pendant group is not involved with the specific Watson-Crick hybridization of the oligonucleotide or oligonucleotide analog with the MRNA but is carried along by the oligonucleotide or oligonucleotide analog to serve as a reactive functionality. The pendant group is intended to interact with the mRNA in some manner more effectively to inhibit translation of the mRNA into protein. Such pendant groups have also been attached to molecules targeted to either single or double stranded DNA. Such pendant groups include, intercalating agents, cross-linkers, alkylating agents, or coordination complexes containing a metal ion with associated ligands. A discussion of pendant groups is set forth in PCT Application assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA Activity and Gene Expression," PCT Patent Application Number PCT\US91\00243, International Publication Number WO 91/10671, the disclosures of which are incorporated herein by reference in order to disclose more fully such modifications.

Prior approaches using cross-linking agents, alkylating agents, and radical generating species as pendant groups on oligonucleotides for antisense diagnostics and therapeutics have several significant shortcomings. The sites of attachment of the pendant groups to oligonucleotides play an important, yet imperfectly known, part in the effectiveness of oligonucleotides for therapeutics and diagnostics. Prior workers have described most pendant groups as being attached to a phosphorus atom which affords oligonucleotide compositions with inferior hybridization properties. Prior attempts have been relatively insensitive, that is the reactive pendant groups have not been effectively delivered to sites on the messenger RNA molecules for alkylation or cleavage in an effective proportion. Moreover, even if the reactivity of such materials were perfect, (i.e., if each reactive functionality were to actually react with a messenger RNA molecule), the effect would be no better than stoichiometric. That is, only one mRNA molecule would be inactivated for each oligonucleotide molecule. It is also likely that the non-specific interactions of oligonucleotide compositions with molecules other then the target RNA, for example with other molecules that may be alkylated or which may react with radical species, as well as self-destruction, not only diminishes the diagnostic or therapeutic effect of the antisense treatment but also leads to undesired toxic reactions in the cell or in vitro. This is especially acute with the radical species that are believed to be able to diffuse beyond the locus of the specific hybridization to cause undesired damage to non-target materials, other cellular molecules, and cellular metabolites. This perceived lack of specificity and stoichiometric limit to the efficacy of such prior alkylating agent and radical generating-types of antisense oligonucleotide compositions is a significant drawback to their employment.

Reactive functionalities or pendant groups attached to oligonucleotide compositions previously described in the literature have been almost exclusively placed on a phosphorus atom, the 5-position of thymine, and the 7-position of purines. A phosphorus atom attachment site can allow a reactive group access to both the major and minor grooves. However, internal phosphorus modification results in greatly reduced heteroduplex stability. Attachments at the 3' and/or 5' ends are limiting in that only one or two functional groups can be accommodated in the oligonucleotide compositions. Such placement can interfere with Watson-Crick binding. Further, functionalities placed in the 5-position or 7-position of bases, pyrimidine and purine, respectively will reside in the major groove of the duplex and will not be in proximity to the RNA 2'-hydroxyl substrate. The 2'-hydroxyl is the "trigger" point for RNA inactivation, and thus, any reactive functionalities must be in appropriate proximity to the receptive substrate located in the targeted RNA, especially the most sensitive point, the 2'-hydroxyl group.

Targeted RNA is inactivated by formation of covalent links between a modified oligonucleotide and the RNA 2'-hydroxyl group. A variety of structural studies such as X-ray diffraction, chemical reaction, and molecular modeling studies suggests that the 2'-hydroxyl group of RNA in a duplex or heteroduplex resides in the minor groove.

The half life of the perfectly formed duplex will be greatly effected by the positioning of the tethered functional group. Inappropriate positioning of functional groups, such as placement on the Watson/Crick base pair sites, would preclude duplex formation. Other attachment sites may allow sequence-specific binding but may be of such low stability that the reactive functionality will not have sufficient time to initiate RNA disruption.

Approaches to perfect complementation between modified oligonucleotides or oligonucleotides and targeted RNA will result in the most stable heteroduplexes. This is desired because the heteroduplex must have a sufficient half life to allow the reactive or non-reactive functionalities of this invention to initiate the cleavage or otherwise disruption of RNA function. The minor side or minor groove of the duplexes formed between such oligonucleotides or modified oligonucleotides and the targeted RNA has been found to be the greatly preferred site for functional group activity.

Therefore, functionalities placed on sequence-specific oligonucleotide compositions (via modified nucleosides) should preferably reside in the minor groove formed between the oligonucleotide composition and the targeted RNA, not interfere with duplex formation or stability, and initiate cleavage or disruption of the RNA. Accordingly, there remains a great need for antisense oligonucleotide compositions that are capable of improved specificity and effectiveness both in binding and in mRNA modulation or inactivation without the imposition of undesirable side effects.

It has now been found that certain positions on the nucleosides of double stranded nucleic acids are exposed in the minor groove and may be substituted without affecting Watson-Crick base-pairing or duplex stability. Reactive or non-reactive functionalities placed in these positions can best initiate cleavage and destruction of targeted RNA or interfere with its activity.

The functionalities point of attachment to the base units, which in turn may be converted to modified oligonucleotide, is critical in the design of compositions for sequence-specific destruction or modulation of targeted RNA. The functionalities must not interfere with Watson-Crick base pair hydrogen bonding rules, as this is the sequence-specific recognition/binding factor essential for selection of the desired RNA to be disrupted. Further, the functionalities should improve the oligonucleotides compositions' pharmacokinetic and/or pharmacodynamic properties, as well as the oligonucleotide compositions' transport properties across cellular membranes. The present invention addresses these, as well as other, needs by presenting novel oligonucleotide intermediates based on the core structure of 3-deazapurines.

The synthesis of the 3-deazaguanine core is known, Cook, et al., J. Am. Chem. Society 1975, 97, 2916; Cook et al., J. Med. Chem. 1978, 21, 1212. 3-deazaguanine is a potent guanine antimetabolite with significant antitumor, antiviral, antibacterial and antiparasitic activities. The corresponding nucleoside, 2'-deoxy-3-deazaguanosine, has exhibited a wide spectrum, Revankar, et al., J. Med. Chem., 27, 1389, 1984, of antiviral and antitumor activity in addition to antibacterial activity against E. coli, Burman, et al., Chem. Scripta 1986, 26, 15. Workers have made certain modifications to 3-deazaguanine (6-aminoimidazo [4,5-c]pyridine) and the corresponding nucleoside 2'-deoxy-3-deazaguanosine (6-amino-1-(2-deoxy-β-D-erythro-pentofuranosyl)) imidazo [4,5-c]pyridin-4(5H)-one resulting in a wide modulation of the heterocyclic ring system's biological activity. See, e.g., Hartman, et al., J. Labelled Compd. Radiopharm. 1985, 23, 35 (ring modifications); Revankar, et al., J. Med. Chem. 1984, 27, 1389 (peripheral modifications); Cook, et al., J. Org. Chem. 1978, 43, 289 (same); Revankar, supra (sugar modifications). Workers have attached certain tether functionalities to the 3-position of 3-deaza-adenine. The Chemistry of Heterocyclic Compounds, A. Weissberger, Ed., Imidazole and Derivatives, Part 1, Interscience, N.Y. (1953). However, there has been no investigation into the synthesis of 3-C substituted deazaguanine. The present invention is the first to set forth substitutions at the 3-C position of 3-deazaguanines and 3-deazapurine derivatives.

The bulkiest 3-C deazapurine substituent induces an unnatural 3'-endo/high-anti (-sc) conformation of the nucleoside. This preference for the anti-conformation may make the 2'-deoxy 3-deazapurines of the invention enhanced substrates for viral kineses. Further, substitutions at the C-3 aromatic carbon of the 3-deazapurine ring system are of interest because this influences the heterocycle's range of rotation about the glycosidic bond, Saenger, "Principles of Nucleic Acid Structure," Cantor, C. R., Ed., Springer-Verlag, New York, 1983, potentially modifying biological activity, Saran, et al., Int. J. Quantum Chem. 1984 25, 743; Miles, et al., H.J. Theor. Biol. 1977, 67, 499. Further, lipophilic substituents at this position could change the transport efficiency of heterocyclic bases, heterocyclic base analogs, nucleosides, nucleoside analogs, nucleotides, nucleotide analogs, and oligonucleotides compositions.

This invention presents novel compounds based on the 3-deazapurine ring system that have utility as intermediates for oligonucleotide compositions. This invention also provides novel synthetic methods for the preparation of these compounds employing a mild alkylation procedure on an imidazole precursor to this ring system. In particular, this invention provides nucleosides, nucleoside analogs, nucleotides, nucleotide analogs, heterocyclic bases, and heterocyclic base analogs. These heterocyclic compounds are adapted for placement of the reactive, RNA cleaving moiety or other reactive moiety into the minor groove site of the hybrid structure formed from the RNA and the composition through careful selection of the attachment of the RNA cleaving moieties.

The compounds of the invention possess unique steric properties that result in modified biological activity and better cellular transport properties for oligonucleotide compositions. These attributes make these compositions useful oligonucleotide intermediates.

In one aspect of the invention, the compounds have the formula: ##STR1## wherein G is C or N; R1 is NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; R2 is alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; and Y is H, a nitrogen protecting group, or a sugar moiety.

In certain preferred embodiments, Y is ribose. In a more preferred embodiment, Y is deoxyribose. In another preferred embodiment, Y is a sugar analog, preferably the deoxyribose type.

In certain preferred embodiments, R1 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, R1 is alkenyl having up to about 12 carbon atoms. In still another preferred embodiment, R1 is aralkyl having from about 6 to about 30 carbon atoms.

In other preferred embodiments, Y is ribose or deoxyribose and R1 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, Y is ribose or deoxyribose and R1 is alkenyl having up to about 12 carbon atoms. In yet another preferred embodiment, Y is ribose or deoxyribose and R1 is aralkyl having from about 6 to about 30 carbon atoms.

In certain other preferred embodiments, G is N, R1 is alkyl, R2 is amino, and Y is ribose or deoxyribose. In another preferred embodiment, G is N, R1 is alkenyl, R2 is amino, and Y is ribose or deoxyribose. In other preferred embodiments, G is N, R1 is aralkyl, R2 is amino, and Y is ribose or deoxyribose. In other preferred embodiments, G is N; R1 is 1-methyloctane, 1-propene, phenylmethyl, or napthylethyl; R2 is amino; and Y is ribose or deoxyribose.

In certain preferred embodiments, the RNA cleaving moiety comprises a portion reactive with said RNA. In another preferred embodiment, the RNA cleaving moiety further comprises a tether portion for attaching the reactive portion to the balance of the composition.

Numerous amine protecting groups are known in the art, and can be used, including the allyloxycarbonyl (Alloc), benzyloxycarbonyl (CBz), chlorobenzyloxycarbonyl, t-butyloxycarbonyl (Boc), fluorenylmethoxycarbonyl (Fmoc), isonicotinyloxycarbonyl (i-Noc) groups. (see, e.g., Veber and Hirschmann, et al., J. Org. Chem. 1977, 42, 3286 and Atherton, et al., The Peptides, Gross and Meienhofer, Eds, Academic Press; New York, 1983; Vol. 9 pp. 1-38). For example, it is known that the Boc group can protect an amine group from base and from reducing conditions but that it can be removed with acid.

The invention further provides compositions comprising a sugar and base moiety as discussed above, with the 3' position of the sugar moiety derivatized with an activated phosphate group.

In another aspect of this invention, mixed sequence oligonucleotides incorporating at least one compound as set forth above are presented.

In another aspect of the invention, the compounds have the formula: ##STR2## wherein G is C or N; R3 is H, NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; R4 is NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; and Y is H, a nitrogen protecting group, or a sugar moiety; provided that when R3 is H, R4 is not NH2.

In certain preferred embodiments, Y is ribose. In a more preferred embodiment, Y is deoxyribose. In another preferred embodiment, Y is a sugar analog, preferably the deoxyribose type.

In certain preferred embodiments, R3 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, R3 is alkenyl having up to about 12 carbon atoms. In still another preferred embodiment, R3 is aralkyl having from about 6 to about 30 carbon atoms.

In other preferred embodiments, Y is ribose or deoxyribose and R3 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, Y is ribose or deoxyribose and R3 is alkenyl having up to about 12 carbon atoms. In yet another preferred embodiment, Y is ribose or deoxyribose and R3 is aralkyl having from about 6 to about 30 carbon atoms.

In certain other preferred embodiments, G is N, R3 is alkyl, R4 is NH2, and Y is ribose or deoxyribose. In another preferred embodiment, G is N, R3 is alkenyl, R4 is NH2, and Y is ribose or deoxyribose. In other preferred embodiments, G is N, R3 is aralkyl, R4 is NH2, and Y is ribose or deoxyribose.

In certain preferred embodiments, the RNA cleaving moiety comprises a portion reactive with said RNA. In another preferred embodiment, the RNA cleaving moiety further comprises a tether portion for attaching the reactive portion to the balance of the composition.

Numerous amine protecting groups are known in the art, and can be used, including the allyloxycarbonyl (Alloc), benzyloxycarbonyl (CBz), chlorobenzyloxycarbonyl, t-butyloxycarbonyl (Boc), fluorenylmethoxycarbonyl (Fmoc), isonicotinyloxycarbonyl (i-Noc) groups. (see, e.g., Veber and Hirschmann, et al., J. Org. Chem. 1977, 42, 3286 and Atherton, et al., The Peptides, Gross and Meienhofer, Eds, Academic Press; New York, 1983; Vol. 9 pp. 1-38). For example, it is known that the Boc group can protect an amine group from base and from reducing conditions but that it can be removed with acid.

The invention further provides compositions comprising a sugar and base moiety as discussed above, with the 3' position of the sugar moiety derivatized with an activated phosphate group.

In another aspect of this invention, mixed sequence oligonucleotides incorporating at least one compound as set forth above are presented.

In another aspect of the invention, novel methods for synthesizing the compounds of the invention are provided. The synthesis of the 3-deazaguanine ring system has been most widely achieved via the base-catalyzed cyclization of a 5(4)-cyanomethyl-imidazole-4(5)-carboxamide. Cook, et al., J. Am. Chem. Soc. 1975, 97, 2916; Cook, et al., R. K. T. Med. Chem. 1978, 21, 1212, the disclosures of which are incorporated herein by reference in their entirety. Both the 2'-deoxy- and the 3-deazaguanosines have also been synthesized using a synthetic scheme that relies on this novel cyclization.

Generally, the compounds of the invention may be synthesized by employing a mild alkylation on the methylene carbon of an imidazole precursor to the 3-deazapurine ring system. The starting material may be a 5-cyano-methyl-imidazole carboxylate, which can be synthesized according to a known procedure. Journal of Medicinal Chemistry, Vol. 27 p. 1389 (1984). For nucleoside synthesis, a sugar moiety is attached to the 1-position of the imidazole ring of the starting material before the alkylation step. Methods of attaching a sugar moity to the imidazole ring are well known to those skilled in the art.

For base synthesis, the ester functionality of the starting material, 5 cyanoimidazole carboxylate, is protected with an ester protecting group before the alkylation step. Any ester protecting groups and methods of attaching ester protecting groups known in the art may be used.

Generally, reactive functionalities emanating from the 3-position of the 3-deazapurine ring can be obtained by a multi-step synthesis under the following reaction conditions. The methylene moiety of the staring material is alkylated with halogenated reactive functionalities, such as an alkyl, alkenyl, or aryl halide.

The alkylated carboxylates are converted to the corresponding carboxamides by ammonolysis at elevated temperature. The alkylated carboxamides are subsequently cyclized using one of several available procedures. Cook, et al., J. Am. Chem. Soc. 1975, 97, 2916; Cook, et al., R. K. J. Med. Chem. 1978, 21, 1212. For example, cyclized products can be obtained using a two-step procedure employing liquid and methanolic ammonia. The protecting groups are removed from the imidazole intermediate with hydrochloric acid.

The corresponding nucleotides of the invention can be prepared by protecting the 5' position of the sugar moiety of a nucleosidic unit and derivatizing the 3' position with an appropriate phosphoramidite or other activated phosphate. These are inserted into oligonucleotides as 5'-DMT-3'-cyanoethyl phosphoramidites through routine solid state synthetic techniques.

Oligonucleotide or oligonucleotide analogs incorporating at least one of the novel compounds of the invention may be synthesized and are within the ambit of this invention. Oligonucleotides or oligonucleotide analogs incorporating the novel compounds of the invention may be synthesized, conveniently through solid state synthesis of known methodology, to be complementary to or at least to be specifically hybridizable with the preselected nucleotide sequence of the RNA or DNA. Nucleic acid synthesizers are commercially available and their use is generally understood by persons of ordinary skill in the art as being effective in generating nearly any oligonucleotide of reasonable length which may be desired. See, e.g., Caruthers, Oligonucleotides, Antisense Inhibitors of Gene Expression., pp. 7-24, J. S. Cohen, ed. (CRC Press, Inc. Boca Raton, Fla., 1989); J. Am. Chem. Society, 1990, 112, 1253-1255; Beaucage et al., Tetrahedron Letters, 1981, 22, 1859-1862.

The sugar moiety of the nucleosidic units incorporated onto the oligonucleotide compositions is preferably the deoxyribose type. The groups linking the bases together may be the usual sugar phosphate nucleic acid backbone, but may also be modified as a phosphorothioate, methylphosphonate, or phosphate alkylated moiety to further enhance the sugar modified oligonucleotide properties, along with removal of a 5'-methylene group and/or carbocyclic sugar. Sugar modifications are disclosed as set forth in PCT Application Number PCT\US91\00243 assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA Activity and Gene Expression," International Publication Number WO 91/10671, the disclosures of which are incorporated herein by reference in order to disclose more fully such modifications.

This invention presents novel heterocyclic compounds based on the 3-deazapurine core that may be used intermediates for oligonucleotide compositions. In particular, this invention provides nucleosides, nucleoside analogs, nucleotides, nucleotide analogs, heterocyclic bases, and heterocyclic base analogs. The compounds of the invention possess unique steric properties that result in modified biological activity and better cellular transport properties. These attributes make these compositions useful intermediates for oligonucleotide compositions.

The compounds of the invention may have at least one RNA cleaving moiety or other moiety capable of interacting with an RNA appended thereto, which are adapted for placement of the reactive RNA cleaving moiety or other reactive moiety into the minor groove site of the hybrid structure formed from the RNA and the composition through careful selection of the sites of attachment of the RNA cleaving moieties. Incorporation of these novel compounds into oligonucleotides compositions improves these compositions' pharmacokinetic and pharmacodynamic properties, the compositions' resistance to nucleases, improves the compositions' binding capabilities without any concomitant interference with the Watson-Crick binding, and enhances the compositions' penetration into cells.

The functionalized sites on the base units, which in turn may be incorporated into modified oligonucleotides, is critical in the design of compositions for sequence-specific destruction or modulation of targeted RNA. The functionality must not interfere with Watson-Crick base pair hydrogen bonding rules as this is the sequence-specific recognition/binding factor essential for selection of the desired RNA to be disrupted.

It has now been found that certain positions on the nucleosides of double stranded nucleic acids are exposed in the minor groove and may be substituted without affecting Watson-Crick base-pairing or duplex stability. Reactive or non-reactive functionalities placed in these positions in accordance with this invention can best initiate cleavage and destruction of targeted RNA or interfere with its activity. The sites of functionality in the heterocyclic compounds of the invention are novel, and have been preferably designed such that the functionalities will reside in or on the minor groove formed by the heteroduplex between modified oligonucleotides and targeted RNA.

The present invention also provides novel methods for the synthesis of the compounds of the invention employing a mild alkylation procedure on an imidazole precursor to the 3-deazapurine ring system.

The compounds possessing the required functionality in the heterocyclic base portion may be used to prepare desired oligonucleotides and oligonucleotide analogs. These oligonucleotide and oligonucleotide analogs are also within the ambit of this invention. Oligonucleotides and oligonucleotide analogs incorporating the novel compounds of the invention are believed to increase the oligonucleotide compositions' nuclease resistance, and thus, facilitate anti-sense therapeutic, diagnostic use, or research reagent use of these antisense agents.

In the context of this invention, a "nucleoside" is a nitrogenous heterocyclic base linked to a pentose equivalent, either a ribose, deoxy ribose, or derivative or analog thereof. The term "nucleotide" means a phosphoric acid ester of a nucleoside comprising a nitrogenous heterocyclic base, a pentose equivalent, and one or more phosphate or other backbone forming groups; it is the monomeric unit of an oligonucleotide. The term "oligonucleotide" refers to a plurality of joined nucleotide units formed in a specific sequence from naturally occurring heterocyclic bases and pentofuranosyl equivalent groups joined through phosphodiester or other backbone forming groups. Nucleotide units may be nucleic acid bases such as guanine, adenine, cytosine, thymine, or derivatives thereof. The pentose equivalent may be deoxyribose, ribose, or groups that substitute therefore. This term refers to both naturally occurring and synthetic species formed or derived from naturally occurring subunits. The terms "antisense agents" and "oligonucleotide compositions" as used in the context of this invention encompass oligonucleotides and oligonucleotide analogs. In the context of this invention, "activated phosphate group" means phosphorothioates, methyl phosphonates, phosphoramidites, and phosphorotriesters (phosphate methylated DNA) and any other groups known to those skilled in the art.

"Modified base," "base analog," "modified nucleoside," "nucleotide analog," or "modified nucleotide," in the context of this invention refer to moieties that function similarly to their naturally occurring counterparts but have been functionalized to change their properties.

"Sugar moiety" as used in the context of this invention refers to naturally occurring sugars, such as ribose or deoxyribose, and sugars and non-sugar analogs that have been functionalized to change their properties.

"Oligonucleotide analogs" or "modified oligonucleotides" as used in connection with this invention, refer to compositions that function similarly to natural oligonucleotides but that have non-naturally occurring portions. Oligonucleotide analogs or modified oligonucleotides may have altered sugar moieties, altered bases, both altered sugars and bases or altered inter-sugar linkages, for example phosphorothioates and other sulfur containing species which are known for use in the art.

In the context of the invention, "improved pharmacodynamic property" means improved oligonucleotide uptake, enhanced oligonucleotide resistance to degradation, and/or strengthened sequence-specific hybridization with RNA. "Improved pharmacokinetic property" means improved oligonucleotide uptake, distribution, metabolism or excretion.

In one aspect of the invention, the compounds have the formula: ##STR3## wherein G is C or N; R1 is NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; R2 is alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; and Y is H, a nitrogen protecting group, or a sugar moiety.

In certain preferred embodiments, Y is ribose or deoxyribose. In a more preferred embodiment, Y is deoxyribose. In another preferred embodiment, Y is a sugar analog, preferably the deoxyribose type. Sugar analogs with substituents at the 3' or 5' of deoxyribose, or at the 2', 3', or 5' of ribose are contemplated. Suitable substituents on the sugar moiety include, but are not limited to, O, H, lower alkyl, substituted lower alkyl, aralkyl, heteroaralkyl, heterocycloalkyl, amino-alkylamino, heterocycloalkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, SOMe, SO2 Me, ONO2, NO2, N3, NH2, NH-alkyl, OCH2 CH═CH2, OCH═CH2, OCH2 CCH, OCCH, or an RNA cleaving moiety.

Generally, substituted sugars may be synthesized according to the methods disclosed in PCT Patent Application Number PCT\US91\00243 assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA Activity and Gene Expression," the disclosures of which are incorporated herein by reference to disclose more fully such modifications.

For example, a substituted sugar as, methyl 3-O-(t-butyldiphenylsilyl)-2,5-dideoxy-5-C-formyl-α/β-D-erythro -pentofuranoside, can be prepared by modifying 2-deoxy-D-ribose to methyl 2-deoxy-α/β-D-erythro-pentofuranoside (prepared according to the method of M.S. Motawai and E.B. Pedersen, Liebigs Ann. Chem. 1990, 599-602), which on selective tosylation followed by 3-O-silylation gave methyl 3-O-(t-butyldimethylsilyl)-2-deoxy-5-O-tosyl-α/β-D-erythro-pent ofuranoside.

In certain other preferred embodiments, R1 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, R1 is alkenyl having up to about 12 carbon atoms. In still another preferred embodiment, R1 is aralkyl having from about 6 to about 30 carbon atoms. In another preferred embodiment, R1 is 1-methyloctane. In a more preferred embodiment, R1 is 1-propene. In still a more preferred embodiment, R1 is phenylmethyl. In yet a more preferred embodiment, R1 is napthylethyl.

In other preferred embodiments, Y is ribose or deoxyribose and R1 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, Y is ribose or deoxyribose and R1 is alkenyl having up to about 12 carbon atoms. In yet another preferred embodiment, Y is ribose or deoxyribose and R1 is aralkyl having from about 6 to about 30 carbon atoms.

In certain other preferred embodiments, G is N, R1 is alkyl, R2 is amino, and Y is ribose or deoxyribose. In another preferred embodiment, G is N, R1 is alkenyl, R2 is amino, and Y is ribose or deoxyribose. In other preferred embodiments, G is N, R1 is aralkyl, R2 is amino, and Y is ribose or deoxyribose. In other preferred embodiments, G is N; R1 is 1-methyloctane, 1-propene, phenylmethyl, or napthylethyl; R2 is amino; and Y is ribose or deoxyribose.

R1 and/or R2 may be any of the following alkyl, alkenyl, aryl, amino, or cyclic groups. Alkyl groups of the invention include, but are not limited to, C1 -C12 straight and branched chained alkyls such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl, isopropyl, 2-butyl, isobutyl, 2-methylbutyl, isopentyl, 2-methyl-pentyl, 3-methylpentyl, 2-ethylhexyl, 2-propylpentyl. Alkenyl groups include but are not limited to unsaturated moieties derived from the above alkyl groups including but not limited to vinyl, allyl, crotyl, propargyl. Aryl groups include but are not limited to phenyl, tolyl, benzyl, naphthyl, anthracly, phenanthryl, and xylyl. Suitable heterocyclic groups include but are not limited to imidazole, tetrazole, triazole, pyrrolidine, piperidine, piperazine and morpholine. Amines include amines of all of the above alkyl, alkenyl and aryl groups including primary and secondary amines and "masked amines" such as phthalimide. Amines are also meant to include polyalkylamino compounds and aminoalkylamines such as aminopropylamine and further heterocycloalkylamines such as imidazol-1, 2 or 4-yl-propylamine. Substituent groups for the above include but are not limited to other alkyl, haloalkyl, alkenyl, alkoxy, thioalkoxy, haloalkoxy and aryl groups as well as halogen, hydroxyl, amino, azido, carboxy, cyano, nitro, mercapto, sulfides, sulfones and sulfoxides. Other suitable substituent groups also include rhodamines, coumarins, acridones, pyrenes, stilbenes, oxazolopryidocarbazoles, anthraquinones, phenanthridines, phenazines, azidobenzenes, psoralens, porphyrins and cholesterols. Pendant groups that do not possess reactive functionality, do not initiate chemical reactions, but enhance the oligonucleotide compositions' pharmacodynamic and pharmacokinetic properties may also be used without departing from the spirit of the invention. Such groups include, but are not limited to, alkyl chains, polyamines, ethylene glycols, polyamides, aminoalkyl chains, amphipathic moieties, points for reporter group attachment, and intercalators attached to any of the preferred sites for attachment.

In other preferred embodiments, the RNA cleaving moiety comprises a portion reactive with said RNA. It is believed desirable in accordance with certain preferred embodiments, to attach the RNA cleaving portion to one of the nucleosides forming the subunits of the oligonucleotide compositions' targeting portion.

In another preferred embodiment, the RNA cleaving moiety further comprises a tether portion for attaching the reactive portion to the balance of the composition. It is not necessary to tether more than one, or perhaps two RNA cleaving functionalities to oligonucleotide compositions in accordance with this invention in order to provide the benefits of the invention. Thus, an RNA cleaving moiety will preferably be tethered to a relatively small proportion of the subunits, generally only one or two, comprising the oligonucleotide compositions, which is the targeting portion of the compositions of the invention. In other embodiments, of the invention, however, all of the nucleotides in an oligonucleotide can be modified to include one or more RNA cleaving moiety groups.

The half life of the perfectly formed duplex will be greatly effected by the positioning of the tethered functional group. Inappropriate positioning of functional groups, such as placement on the Watson/Crick base pair sites, would preclude duplex formation. Other attachment sites may allow sequence-specific binding but may be of such low stability that the reactive functionality will not have sufficient time to initiate RNA disruption.

It is believed that attaching RNA cleaving moieties in accordance with the foregoing considerations will permit those moieties to lie in the minor groove of the hybrid formed from the composition of the present invention and the messenger RNA for which modulation is desired. It is possible that other positions for attachment of the RNA cleaving moieties having a similar effect may be found, especially when further modifications of the purine structure is undertaken as may be done by persons of ordinary skill in the art without deviating from the spirit of the present invention. Once again, it is to be understood that preferably one, or at most a few RNA cleaving moieties are generally to be employed. Thus, artisans in the field will have great latitude in selecting means of attachment of the RNA cleaving moieties, the pharmacodynamic improving group or the pharmacokinetic improving group in accordance with this invention.

The RNA cleaving moieties of the compositions of the present invention are designed in such a fashion that they can be effective in performing their proximate task, leading to the desired modulation of RNA activity. RNA cleaving moieties may include heteroatomic substitutions; these heteroatomic substituents include, but are not limited to, amides and polyamides, and heterocyclics, especially imidazoles and other nitrogen heterocycles.

Generally, protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as amine groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. See, e.g., Greene and Wuts, Protective Groups in Organic Synthesis, 2d edition, John Wiley & Sons, New York, 1991. Numerous amine protecting groups are known in the art, including, but not limited to, the allyloxycarbonyl (Alloc), benzyloxycarbonyl (CBz), chlorobenzyloxycarbonyl, t-butyloxycarbonyl (Boc), fluorenylmethoxycarbonyl (Fmoc), isonicotinyloxycarbonyl (i-Noc) groups. (see, e.g., Veber and Hirschmann, et al., J. Org. Chem. 1977, 42, 3286 and Atherton, et al., The Peptides, Gross and Meienhofer, Eds, Academic Press; New York, 1983; Vol. 9 pp. 1-38). For example, it is known that the Boc group can protect an amine group from base and from reducing conditions but that it can be removed with acid. Other nitrogen protecting groups will be apparent to those skilled in the art and may be used without detracting from the spirit of the invention.

Another aspect of the invention presents compounds having the formula: ##STR4## wherein G is C or N; R3 is H, NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; R4 is NH2, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aralkyl, amino, amino alkylamino, aralkylamino, substituted alkylamino, heterocycloalkyl, heterocycloalkylamino, aminoalkylamino, hetrocycloalkylamino, polyalkylamino, or an RNA cleaving moiety; and Y is H, a nitrogen protecting group, or a sugar moiety; provided that when R3 is H, R4 is not NH2.

In certain preferred embodiments, Y is ribose or deoxyribose. In a more preferred embodiment, Y is deoxyribose. In another preferred embodiment, Y is a sugar analog, preferably the deoxyribose type. Sugar analogs with substituents at the 3' or 5' of deoxyribose, or at the 2', 3', or 5' of ribose are contemplated. Suitable substituents on the sugar moiety include, but are not limited to, O, H, lower alkyl, substituted lower alkyl, aralkyl, heteroaralkyl, heterocycloalkyl, amino-alkylamino, heterocycloalkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, SOMe, SO2 Me, ONO2, NO2, N3, NH2, NH-alkyl, OCH2 CH═CH2, OCH═CH2, OCH2 CCH, OCCH, or an RNA cleaving moiety. The discussion of sugar moieties set forth above in connection with the other compounds of the invention is fully applicable here.

In certain preferred embodiments, R3 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, R3 is alkenyl having up to about 12 carbon atoms. In still another preferred embodiment, R1 is aralkyl having from about 6 to about 30 carbon atoms. In a more preferred embodiment, R3 is 1-methyloctane. In a more preferred embodiment, R3 is 1-propene. In still a more preferred embodiment, R3 is phenylmethyl. In yet a more preferred embodiment, R3 is napthylethyl.

In other preferred embodiments, Y is ribose or deoxyribose and R3 is alkyl having up to about 12 carbon atoms. In another preferred embodiment, Y is ribose or deoxyribose and R3 is alkenyl having up to about 12 carbon atoms. In yet another preferred embodiment, Y is ribose or deoxyribose and R3 is aralkyl having from about 6 to about 30 carbon atoms.

In certain other preferred embodiments, G is N, R3 is alkyl, R4 is NH2, and Y is ribose or deoxyribose. In another preferred embodiment, G is N, R3 is alkenyl, R4 is NH2, and Y is ribose or deoxyribose. In other preferred embodiments, G is N, R3 is aralkyl, R4 is NH2, and Y is ribose or deoxyribose. In other preferred embodiments, G is N; R3 is 1-methyloctane, 1-propene, phenylmethyl, or napthylethyl; R4 is amino; and Y is ribose or deoxyribose.

R3 and R4 may be any of the alkyl, alkenyl, aryl, amino, or cyclic groups as set forth above.

In other preferred embodiments, the RNA cleaving moiety and nitrogen protecting groups may be as set forth above in connection with the previously discussed compounds. The discussion set forth above is fully applicable here.

Generally, the bases of the invention may be synthesized by starting with a 5-cyanomethyl imidazole carboxylate, protecting the ester with an ester protecting group, treating the compound with sodium hydride, followed by electrophilic substitution with an alkyl, alkenyl, or aryl halide, preferably bromine. The carboxylates are converted to the corresponding carboxamides by treatment with methanolic ammonia at elevated temperature. The alkylated products are subsequently cyclized. Cook, et al., J. Am. Chem. Soc. 1975, 97, 2916; Cook, et al., R. K. J. Med. Chem. 1978, 21, 1212; Revankar, supra.

The following discussion provides an illustrative example of a possible synthetic route. The bases of the invention may be synthesized by protecting the methyl 5(4)-cyanomethyl-imidazole-4(5)-carboxylate's imidazole ring nitrogens with a protecting group such as a tetrahydropyranyl group. A reaction with 2,3-dihydropyran in the presence of tosic acid yields the 5 and 7 tetrahydro-pyranyl positional isomers of 4-cyanomethyl-imidazole-5-carboxylates in roughly a 2:1 ratio. These isomers are separated in order to isolate the isomer with a tetrahydropyranyl protecting group in a position removed from the proposed site of alkylation. Cook et al., J. Med. Chem., 1978, 21, 1212.

The 7 tetrahydropyranyl positional isomer is then alkylated by adding a dilute solution of the alkylating electrophile in acetonitrile to the reaction mixture, the reaction mixture is stirred under an inert atmosphere for periods of 3-18 hours. The reaction media and products are isolated by flash-column chromatography using silica gel. In each case, the products are approximately 1:1 mixtures of isomers, as determined by an integration of the signals for the H-2 protons in the 1 H-NMR. The alkylated product mixtures were subsequently treated with methanolic ammonia and heated at 75°C in a sealed vessel to yield the cyclized products. When the ammonolysis was conducted at 100°C, there was extensive decomposition and no products could be isolated. The mixtures were evaporated under reduced pressure to afford light colored solids which decomposed rapidly when exposed to air and moisture. The crude reaction products were thoroughly rid of all ammonia and treated with 1N HCl in methanol for several hours to remove the tetrahydropyranyl protecting groups, which yielded the 7-alkyl (aryl)-6-amino-1,5-dihydroimidazo [4,5-c]pyridin-4-one products as stable hydrochloride salts. Hartman, et al., J. Labelled Compd. Radiopharm., 1985, 23, 35. The hydrochloride salts gave satisfactory C,H,N analyses and 1 H-NMR spectra after months of storage, attesting to their long-term stability. The following reaction scheme illustrates this synthesis. ##STR5##

Compounds of the invention having a sugar moiety at the 1-position of the imidazole ring invention may be synthesized using a procedure similar to that used for the previously discussed bases of the invention. Generally, reactive functionalities emanating from the 3-position of a 2'-deoxy-3-deazapurine can be obtained by a multi-step synthesis starting with the alkylation of the methylene moiety of a cyanomethyl imidazole derivative with various electrophiles, such as halogenated reactive functionalities. The alkylated product is ammonylated to yield the carboxamide. The carboxamide of methyl-5-(cyanomethyl)-1-(2'deoxy-3,5-di-O-p-toluyl-β-D-erythro-pento furanosyl) imidazole-4-carboxylate may be obtained from the ammonolysis of the methyl 5-cyanomethyl-imidazole-4-carboxylates, and 5-cynanomethyl-1-(2-deoxy-3,5-di-O-p-toluoylp-β-D-erythro-pentofurano syl)-imidazole-4-carboxylate. Cyclized products may be obtained using a two-step procedure employing liquid and then methanolic ammonia. The alkylated imidazoles are treated with methanolic ammonia to remove the toluoyl groups from the sugar moiety. These may be inserted into oligonucleotides as 5'-DMT-3'-cyanoethyl phosphoramidites through routine solid state synthetic techniques.

The starting imidazole material, 5-cyano (substituted)-methyl-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pento furanosyl) imidazole-4-carboxylate, for the above discussed reaction, can be synthesized according to a known procedure. Journal of Medicinal Chemistry, Vol. 27 p. 1389 (1984). The synthetic intermediate methyl-5-(cyanomethyl)-1-(2'deoxy-3,5-di-O-p-toluyl-β-D-erythro-pento furanosyl) imidazole-4-carboxylate was obtained from the sodium salt glycosylation of methyl 5(4)-cyanomethyl-imidazole-4(5)carboxylate with 1-chloro-2-deoxy-3,5-di-O-p-toluoyl-a-D-erythro-pentofuranose (chlorosugar) in acetonitrile. Positional isomers corresponding to methyl 5-cynanomethyl-1-(2-deoxy-3,5-di-O-p-toluoylp-β-D-erythro-pentofurano syl)-imidazole-4-carboxylate and methyl 4-cyanomethyl-1-(2-deoxy-3,5-di-O-p-toluoyl-b-D-erythro-pentofuranosyl)-im idazole 5-carboxylate were obtained in 1:1.5 ratios and in good overall yields. The most favorable ratio of positional isomers was obtained when the chloro-sugar used in the condensation was free of acidic contaminants and anhydrous conditions were maintained during the course of the reaction. Revankar, supra. Other synthetic methods will be apparent to those skilled in the art.

The nucleosides may be synthesized under the following reaction conditions. The 5-cynanomethyl-1-(2-deoxy-3,5-di-O-p-toluoylp-β-D-erythro-pentofurano syl)-imidazole-4-carboxylate was equilibrated with an excess of sodium hydride at room temperature in acetonitrile. A dilute solution of the alkylating agent in acetonitrile was introduced into the reaction mixture and the reaction mixture stirred under an inert atmosphere for a period of 3-18 hours. The reaction media and products are then isolated by flash-column chromatography. This procedure yields mixtures of diastereomeric products. For example, in the case of the ethyl 5-(benzyl[cyano]methyl) imidazole-4-carboxylate or the methyl 5-(cyano[2-(1-naphthyl)ethyl]methyl)imidazole-4-carboxylate 2'-deoxynucleosides, these mixtures were resolved according to the procedures specified in Cook, J. Med. Chem., supra.

For example, in the case of the methyl 5-(benzyl [cyano]methyl)imidazole-4-carboxylate or the methyl 5-(cyano [2-(1-naphthyl)ethyl]methyl)imidazole-4-carboxylate 2'-deoxynucleosides, these mixtures were resolved and the diastereomers characterized individually by 1 H-NMR. In these cases, each isomer exhibited a methene proton resonance as a doublet of doublets at δ,4.8 to 5.5 ppm, indicative of a tertiary proton, Pretsch, supra, as part of an AM2 system. In addition, these compounds exhibited nitrile absorption bands at 2220 to 2240 cm-1 in their infrared spectra.

The alkylation products, methyl 5-(cyano[alkyl]methyl)-1-(2-deoxy-3,5di-O-p-toluoyl-β-D-erythro-pento furanosyl) imidazole-4-carboxylate nucleosides are heated in liquid ammonia at 90°C for 18-20 hours in a stainless steel bomb to yield the corresponding deprotected carboxamides. Further heating of the carboxamides with methanolic ammonia at 95°C yields the desired nucleoside products, 2'-deoxy-3-alkyl(aryl)-3-deazapurines. This two step procedure resulted in better yields of the cyclized products than obtained by prolonged heating of the methyl 5-cyanomethyl-imidazole-4-carboxylates in liquid ammonia. Cook, J. Am. Chem. Society, supra. The following reaction scheme illustrates the synthesis of the nucleosides of the invention. ##STR6##

The electrophiles suitable for practicing the invention include, but are not limited to, alkyl, alkenyl, aryl, amino, cyclic, or heterocyclic compounds having a group. The suitable leaving groups include, but are not limited to, chloride, flouride, iodine, and bromine, with bromine preferred.

Any ester protecting groups known to those skilled in the art may be used; tetrahydropyranyl is an example of such a group. However, various ester protecting groups are known and may be used without detracting from the spirit of the invention. See Green and Wuts, spura.

Methods of synthesizing compounds where the 3 position of the imidazole ring system is carbon rather than nitrogen will be apparent to those skilled in the art and may be used without detracting from the spirit of the invention. See Revankar, supra.

Methods of attaching sugar moieties to the imidazole ring are well known in the art. See Revankar, supra.

In preparing certain of the compounds of the invention fugitive masking groups may be used. Such masking groups allow for ease of synthesis of the compounds. The masking groups are subsequently converted to the desired functionality. Such conversion preferably occurs during a standard deblocking step for a later reaction. As an example of use of this procedure is the use of phthalimide group for the introduction of an amino functionality. Alkyl phthalimides are attached at the proper position in a compound of interest, as for example a nucleoside, via a suitable intermediate such as an N-(haloalkyl)phthalimide. Upon completion of the synthesis of the compound of interest it is then used as a structural nucleotide for oligonucleotide synthesis utilizing standard oligonucleotide synthetic techniques on a nucleotide synthesizer. After the desired oligonucleotide or oligonucleotide analog is completed, it is cleaved from the synthesizer support and in doing so the cleaving reagent also converts the alkylphthalimide to the desired alkylamine. The above procedure can be expanded to attach longer chain polyamino functionalities to the oligonucleotides or oligonucleotide analogs of the invention. Nucleotides, nucleotide analogs, oligonucleotide analogs, or oligonucleotides having a first alkylamino functionality are treated with a further N-(haloalkyl) phthalimide. The extended functionality is then treated to yield the terminal amine group. This can be repeated to further extend the polyamino functionality as desired. Alternately, the extended polyamino functionality is first synthesized and reacted with the first alkylamino functionality to form the polyamino functionality.

If any of the reactions yield a reaction slurry from which the product must be recovered this can be done according to nay methods known in the art. Methods of recovering the sample include any filtration or separation techniques known in the art. Such methods include, but are not limited to, vacuum filtration, separatory extraction, or distillation. A preferred method is filtration using air or liquid, but other methods will be apparent to those skilled in the art.

If any products require further washing with organic solvents to separate impurities, reaction intermediates, or byproducts can be done according to methods known in the art. Suitable organic solvents include, but are not limited to, ether, methanol, ethanol, ethyl acetate, or hexanes. Other types of solvents will be apparent to those skilled in the art. Any organic solvent should be evaporated using methods known in the art. Evaporation methods may be accomplished at room temperature, by vacuum, aspiration, or by using latent heat. The evaporation methods are not limited to these techniques and other techniques will be apparent to those skilled in the art.

The reaction product may require purification. This may be accomplished using purification techniques known in the art. These techniques include, but are not limited to, column chromatography, flash chromatography, recrystillization, or gel chromatography. Flash chromatography on silica gel is preferred but other methods will be apparent to those skilled in the art. Any organic solvents suitable for chromatography may be used. These include, but are not limited to, methanol, acetonitrile, hexanes, carbontrichloride, and ethyl acetate. Other solvents will be apparent to those skilled in the art and may be used without detracting from the spirit of the invention.

Generally, nucleotides of the invention may be prepared by protecting the 5' position of the sugar moiety of the imidazole ring and derivatizing the 3' position with an appropriate phosphoramidite or other activated phosphate suitable for use on a DNA synthesizer.

Alkylation of the imidazole active methylene with α-halo-α-[reactive functionality] acetaldehyde dimethylketal or α-halo-methyl-[reactive functionality] ketone with subsequent amination provides imidazole carboxamides. These can be converted to 5'-DMT-3'-cyanoethylphosphoramidites and inserted into sequence specific oligonucleotides. The prepared oligonucleotides, which are in blocked form, are removed from the solid support upon which they are elaborated such as by ammonium hydroxide treatment. Basic treatment as above removes the oligonucleotides from the solid support and cyclizes the imidazole moiety to a 3-deaza-3-[reactive functionality]-guanine residue with the desired oligonucleotide sequence. Further cyclization between the resulting N-2-exocyclic amine group and the aldehydic or ketonic carbonyl provides tricyclic heterocycle with reactive functionality, (pyrrolo y2,3-β]-imidazo [2,3-δ]pyridin-2-one (5H)-7-(or 8)-[reactive functionality]). Alternately, cleavage from the support with concentrate ammonium hydroxide directly cyclizes the imidazole carboxamide to the 3-deazaguanine.

Direct deoxyribosylation of 6-isobutryl pyrrolo[2,3-δ] imidazo [2,3-δ] pyridine 7-(or 8)-[reactive functionality] provides the 1-(2'-deoxy-β-D-erythro-pentofuransyl) derivative after basic deblocking of the toluoyl groups. The tricyclic heterocycle can be obtained from the alkylation of the tetrahydropyranyl derivative of methyl 5-cyanomethylimidazole 4-carboxylate in accordance with the procedure of the Journal of Medicinal Chemistry, Vol. 21, p. 1212 (1978), with α-halo-α-[reactive functionality]-acetaldehyde dimethyl-ketals or α-halomethyl-[reactive functionality] ketones with subsequent amination. Acid treatment removes the tetrahydropyranyl blocking group and reducing conditions provides the 7,8-di-hydro 7-(or 8)-[reactive functionality] tricyclic heterocycle. The dihydropyrrole ring nitrogen can be protected with an isobutryl group. The 5'-DMT-3'-phosphoramidite-6-isobutryl nucleoside can be inserted into sequence-specific oligonucleotides via standard automated synthesis. Oligonucleotides prepared in this manner contain a pyrrolo[2,3-b]imidazo[2,3-d]-pyridin-4-one (5H)-7-(or 8)-[reactive functionality], the 7,8-dihydro ring replacing a normal guanine residue.

In another aspect of the invention, oligonucleo-tides or oligonucleotide analogs incorporating the novel compounds of the invention are provided. Generally, the oligonucleotides or oligonucleotide analogs may comprise a sugar modified or native oligonucleotide containing a target sequence that is specifically hybridizable with a preselected nucleotide sequence, a sequence of DNA or RNA that is involved in the production of proteins whose synthesis is ultimately to be modulated or inhibited in entirely, of single stranded or double stranded DNA or RNA molecule and which is nuclease resistant.

Oligonucleotides or oligonucleotide analogs incorporating the novel compounds of the invention may be synthesized, conveniently through solid state synthesis of known methodology, to be complementary to or at least to be specifically hybridizable with the preselected nucleotide sequence of the RNA or DNA. Nucleic acid synthesizers are commercially available and their use is generally understood by persons of ordinary skill in the art as being effective in generating nearly any oligonucleotide of reasonable length which may be desired. An oligonucleotide or oligonucleotide analog may then be constructed on a synthesizer incorporating one or more of the 5-cyano-[reactive substituent]-methyl imidazole compounds in its sequence.

The resulting novel oligonucleotides or oligonucleotide analogs are synthesized by the standard solid phase, automated nucleic acid synthesizer such as the Applied Biosystems, Incorporated 380B or MilliGen/Biosearch 7500 or 8800. Triester, phosphoramidite, or hydrogen phosphonate coupling chemistries, M. Caruthers, Oligonucleotides. Antisense Inhibitors of Gene Expression., pp. 7-24, J. S. Cohen, ed. (CRC Press, Inc. Boca Raton, Fla. 1989), are used in with these synthesizers to provide the desired oligonucleotides or oligonucleotide analogs. The Beaucage reagent, Journal of American Chemical Society, Vol. 112, pp. 1253-1255 (1990) or elemental sulfur, S. Beaucage et al., Tetrahedron Letters, Vol. 22, pp. 1859-1862 (1981), is used with phosphoramidite or hydrogen phosphonate chemistries to provide substituted phosphorothioate oligonucleotides.

The oligonucleotides or oligonucleotide analogs may further comprise a reactive portion appended to the novel base portion; this reactive functionality may be attached to the base with a tether group. For example, the oligonucleotide or oligonucleotide analogs may further comprise a reactive portion capable of catalyzing, alkylating, or otherwise effecting the cleavage of RNA, especially of its phosphodiester bonds. This reactive portion may be connected to the targeting portion by a tether. The reactive functionalities may either be basic, acidic, or amphoteric. Heteroatomic species can be formulated to be either basic or acidic or, indeed, to be amphoteric for such purposes. Alkylating and free radical forming functionalities may also be used for these purposes. These functionalities are disclosed as set forth in PCT Application Number PCT\US91\00243 assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA Activity and Gene Expression," the disclosures of which are incorporated herein by reference to disclose more fully such functionalities.

These oligonucleotide compositions comprise a targeting portion specifically hybridizable with a preselected nucleotide sequence of RNA, some of the phosphodiester bonds may be substituted with a structure that functions to enhance the compositions' ability to penetrate into cells' intracellular region where the RNA, whose activity is to be modulated, is located. Such substitutions comprise phosphorothioate bonds, short chain alkyl, cycloalkyl structures, structures that are substantially non-ionic and non-chiral. Phosphodiester bond modifications are disclosed as set forth in U.S. patent applications assigned to a common assignee hereof, entitled "Backbone Modified oligonucleotide Analogs," Ser. No. 703,169 and "Heteroatomic oligonucleotide Linkages," Ser. No. 903,160, the disclosures of which are incorporated herein by reference to disclose more fully such modifications. Backbone modifications may be used without departing from the spirit of the invention.

As will be appreciated by persons of ordinary skill in the art, variations in the structures of the sugar moieties useful in the preparation of the subject compositions may be made without deviating from the spirit of the invention. Suitable substituents on the sugar moiety include, but are not limited to, O,H, lower alkyl, substituted lower alkyl, aralkyl, heteroalkyl, heterocycloalkyl, amino-alkylamino, heterocycloalkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, SOMe, SO2 Me, ONO2, NO2, N3, NH2, NH-alkyl, OCH2 CH═CH2, OCH2 CCH, OCCHO, or an RNA cleaving moiety. Generally, substituted sugars may be synthesized according to methods disclosed in PCT Patent Application Number PCT\US91\00243 assigned to a common assignee hereof, entitled "Compositions and Methods for Detecting and Modulating RNA activity and Gene Expression," the disclosures of which are incorporated herein by reference to fully disclose such modifications. See also Motawai, supra.

Once again, it is not necessary that every sugar linking function be in a modified form a substantial number and even a predominance of such linking groups may exist in he native, phosphodiester form as long as the overall targeting portion of the compositions of the molecules exhibits an effective ability to penetrate into the intracellular spaces of cells of the organism in question or otherwise to contact the target RNA and to specifically bind therewith to form a hybrid capable of detecting and modulating the RNA activity. Of course, fully unmodified, native phosphodiester structure as well.

Modifications that may provide oligonucleotides or analogs that are substantially less ionic than native forms and facilitate penetration of modified or unmodified oligonucleotides into the intracellular spaces are also contemplated by this invention. Any of the existing or yet to be discovered methods for accomplishing this goal may be employed in accordance with the practice of the present invention. As will be appreciated by those skilled in the art, modifications of the phosphate bond find utility in this regard. Variations in the phosphate backbone useful in the preparation of the subject compositions may be made without deviating from the spirit of the invention. Modifications at the phosphorous atom are set forth in an application having U.S. application Ser. No. 558,663 and assigned to a common assignee hereof, said application being entitled "Polyamine Oligonucleotides to Enhance Cellular Uptake," filed Jul. 27, 1990 the modification of the sugar structure including the elimination of one of the oxygen functionalities may permit the introduction of such substantially non-chiral, non-ionic substituents in this position.

Standard backbone modifications such as substituting P for S, Me-P, MeO-P, H2 N-P, etc. These substitutions are thought in some cases to enhance the sugar modified oligonucleotide properties. Such substitutions include, but are not limited to, phosphorothionate, methyl phosphonate, or alkyl phosphate. Backbone modifications are disclosed as set forth in U.S. patent applications assigned to a common assignee hereof, entitled "Backbone Modified Oligonucleotide Analogs," Ser. No. 703,619 and "Heteroatomic Oligonucleotide Linkages," Ser. No. 903,160, the disclosures of which are incorporated herein by 10 reference in order to disclose more fully such modifications. The entirety of the disclosure of these applications are incorporated herein by reference in order to disclose more fully such modifications.

The present invention is further described in the following examples. These examples are for illustrative purposes only, and are not to be construed as limiting the appended claims.

Chromatography and Purification.

Silica gel used for flash chromatography was ICN 60 (Costa Mesa, Calif.), 32-63 mesh. Materials not soluble in the solvent system used for flash chromatography (FC) were co-evaporated onto E. Merck silica gel 100 (Darmstadt, Republic of Germany), 70-230 mesh, using a suitable solvent. The dry materials were then applied to the top of a FC column. TLC was performed on prescored E. Merck Kieselgel 60 F254 plates. Compounds were visualized by illuminating TLC plates under UV light (254 nm) and/or by spraying with 10 methanolic H2 SO4 followed by heating. Evaporations were carried out at 40°-50°C using a rotary evaporator and a vacuum pump coupled to a vacuum controller. 1 H-NMR spectra were obtained at 400 mHz in dmso-d6 unless otherwise noted. Where relevant, treatment of samples with D2 O recorded exchangeable protons. Infrared spectra were recorded on a Perkin-Elmer 16PC FT-IR spectrophotometer. Solvent system A=ethyl acetate-hexanes, 3:2; B=ethyl acetate-methanol, 9:1, v/v.

Synthesis of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate

A large scale synthesis of the methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate, a starting material for this work was carried out according to the sodium-salt glycosylation procedure described in Revenkar, supra.

Synthesis of methyl 5-(cyanomethyl)-3-tetrahydropyranyl-imidazole-4-carboxylate Methyl 5-cyanomethyl-3-tetrahydropyranyl-imidazole-4-carboxylate was prepared and separated from its positional isomer according to the procedure described in Cook, J. Med. Chem., supra.

Synthesis of 2'-deoxy-3-deazaguanosine (6-amino-1-(2-deoxy-β-D-ezythro--pentofuranosyl) imidazo [4,5-c]pyridin-4(5R)-one.

A small scale synthesis of the 2'-deoxy-3-deazaguanosine (6-amino-1-(2-deoxy-β-D-erythro-pentofuranosyl) imidazo[4,5-c]pyridin-4(5H)-one yielded material identical in every respect with that reported in Revenkar, supra. The alkylation procedures described below yield diastereomeric mixtures differing in configuration at the alkylated (methene) carbon. For the nucleosides, these mixtures exhibit well-resolved signals for their H-2, methene, H1' and other protons in their 400 mHz 1 H-NMR spectra.

General Nucleoside Alkylation Method. Methyl 5-(cyano[nonyl]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-eythro-pent ofuranosyl) imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate (5.7 g, 11 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.88 g, 60% in oil, washed with hexanes) at room temperature and under an atmosphere of argon. This suspension was stirred for 15 minutes and then treated with iodononane (7.5 mL, 37.4 mmol) via syringe. The reaction mixture was stirred under these conditions for 6 hr; thin layer chromatography showed the disappearance of starting material nucleoside (Rf=0.45, solvent A) and the appearance of two closely migrating and faster products (Rf=0.65, avg). The reaction was quenched with the addition of glacial acetic acid to pH 5 and then evaporated to dryness in vacuo to afford a yellow syrup. The syrup was redissolved in dichloromethane (150 mL) and the solution was washed with cold 0.1N HCl, water, and then dried over magnesium sulfate. Filtration and evaporation of the organic layer afforded a yellow gum which was purified using FC on silica gel (120 g) using a gradient of ethyl acetate in hexanes (20 to 50%). Fractions corresponding to the alkylated products were pooled and evaporated to yield Methyl 5-(cyano[nonyl]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pen tofuranosyl) imidazole-4-carboxylate as a foam, 2.9 g (47%). 1 H-NMR: δ,8.18 and 8.15 (s,s; 1 H; H-2); 6.48 and 6.37 (t,t; 1 H; H1'); 4.68 and 4.36(2 m, 1 H, methene); 3.32 (s, 3H, COOCH3); 2.05, 1.75, 1.18 and 0.90(4 m; 19 H; nonyl). IR (film): 2240 cm-1 (CN). Anal. Calcd for C37H45N307 (643.778): C, 69.03, H, 7.04, N, 6.53. Found: C, 68.77, H, 6.97, N, 6.39.

The bulkiest 3-C substituent on the 3-deazapurine ring system induces the compounds into an unnatural 3'-endo/high-anti (-sc) conformation. Two-dimensional NMR experiments (2D-NOESY) indicate that substituted 2'-deoxy-3-deazaguanosines prefer the anti-conformation. The bulky substituent in 2'-deoxy-3-(2-[1-naphthyl]ethyl)-3-deazaguanosine appears to force the sugar into an unusual 3'-endo conformation. Gradually decreasing the size of these substituents is expected to increase the equilibrium population of the sugar towards 2'-endo. These structural changes may be correlated with biological activity; it is reasonable to expect that the 2'-deoxy-3-alkyl(aryl)-3-deazapurines might be enhanced substrates for viral kineses due to their preference for the anti-conformation.

The 2'-deoxy-3-alkyl(aryl)-3-deazaguanosines have uv spectra with nearly identical maxima to that of 2'-deoxy-3-deazaguanosine. Also, their proton NMR exhibit H-2 (IUPAC numbering) aromatic resonances at δ7.9 to 8.0 ppm, compared to 7.9 ppm for 2'deoxy-3-alkyl(aryl)-3-deazaguanosines, evidence of the nearly negligible electronic contributions from these alkyl or aryl substitutions to the overall purine ring current. However, the 2D-NOESY NMR spectrum of 2'-deoxy-3-(2-[1-naphthyl] ethyl)-3-deazaguanosine, (7-(2-[1-naphthyl] ethyl)-6-amino-1-(2deoxy-β-D-erythro-pentofuranosyl) imidazo[4,5)-c]pyridin-4(5H)-one, in dmso-d reveals some interesting features. Poonian, supra. The cross-peaks for the imidazole H-2 and sugar H-3'-protons are strong and there are weak cross peaks for the H-2 and H-2' protons. In addition, the H-2" (α) signal appears upfield of the H-2' (β) signal, a configuration not normally registered for 2'-deoxynucleosides. Taken together, these observations indicate that the pseudorotation, Saenger, supra, of the sugar strongly favors 3'-endo (75%) and that this conformation forces the H-2" proton into the shielding volume of the heterocyclic rings. The relative populations of 2' and 3-endo conformations of the nucleosides were determined from their 2D-NOESY spectrum, by measurement of the volume of the cross peaks of H-2 and H-2' (β) versus H-2 and H-3' and divided by the sum of both cross peak volumes. An absence of NOE signals for the N-H imino and 3'-protons indicate that this and other nucleosides in this series appear to exist strictly in an anti conformation. Rosenmeyer, et al., J. Org. Chem., 1990, 55, 5784. Further, because there is a strong NOE signal from H-2 and H-1', it appears that this nucleoside is virtually locked into a high-anti (-sc) conformation. In contrast, the 2D-NOESY spectrum for the 2'-deoxy-3-allyl-3-deazaguanosine (7-allyl-amino-1-(2-deoxy-β-D-erythro-pentofuranosyl) imidazo[4,5c]pyridin-4(5H)-one) exhibits strong cross-peaks for the imidazole H-2 and sugar H-2' protons and lesser cross peaks for H-2 and H-3'1, indicating 2'-endo (62%) as a predominant conformation for the deoxyribose sugar.

Synthesis of Methyl 5-(allyl[cyano]methyl)-1-(2'deozy-3,5-di-O-p-toluoyl-β-D-erythro-pent ofuranosyl) imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate (5.0 g, 9.7 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.46 g, 11.6 mmol) and then allyl bromide (2.5 mL, 29 mmol). Workup of the reaction and purification of the products on silica gel (75 g) as described in Example 1 afforded methyl 5-(allyl[cyano]methyl)-1-(2'deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pent ofuranosyl) imidazole-4-carboxylates a yellowish foam, 3.7 g (68%). 1 H-NMR (200 mHz): δ,8.15 and 8.13 (s,s; 1 H; H-2); 6.38 (m, 1H; H1'); 5.75 and 5.08 (2m; 3H; vinyl); 3.79 (s, 3H, COOCH3). Anal. Calcd for C32 H31 N3 O7 (557.60): C, 25 66.77, H, 5.60, N, 7.53. Found: C, 66.43; H, 5.59, N, 7.38.

Synthesis of Methyl 5-(benzyl[cyano]methyl)-1-(2'-deoxy-3,5 di-o-p-toluoyl-β-D-erythro-pentofuranosyl)imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate (5.0 g, 9.6 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.46 g, 11 mmol) under argon and stirred at room temperature for 15 minutes. The mixture was cooled to 4°C in an ice bath and a solution of benzyl bromide (1.26 mL, 10.6 mmol) in acetonitrile (15 mL) was added dropwise over 70 min. The ice bath was removed and the reaction further stirred at room temperature for 2.5 hours. Workup of the reaction and purification of the products on silica gel (100 g) as described in the Example 1 afforded methyl 5-(benzyl[cyano]methyl)-1-(2'-deoxy-3,5di-O-p-toluoyl-β-D-erythro-pen tofuranosyl) imidazole-4-carboxylate as a white foam, 3.4 g (58%). 1 H-NMR: δ,8.12 and 8.05 (s,s; 1H; H-2); 8.0-7.10 (m, 13 H; aromatic); 6.33 and 6.01 (t,t; 1H; H-1'); 5.22 and 5.02 (t,t; 1H; methene); 3.80 (s, 3H, COOCH3). IR (film): 2240 cm-1 (nitrile). Anal. Calcd for C35 H33 N3 O7 (607.66): C, 69.18; H, 5.47; N, 6.92. Found: C, 69.15; H; 5.43; N, 6.82.

Synthesis of Methyl 5-(cyano[2-(1-naphthyl)ethyl]methyl)-1-(2-deoxy-3,5di-O-p-β-D-erythro -pent imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3, 5-di-O-p-toluoyl-β-D-erythro-pentofuranosyl) imidazole-4-carboxylate (7.43 g, 14.3 mmol) in anhydrous acetonitrile (50 mL) was treated with sodium hydride (1.15 g, 28.7 mmol) under argon and stirred at room temperature for 15 minutes. The 2-(1-naphthyl)ethyl bromide (16.9 g, 71.5 mmol) was added neat and the reaction stirred for 18 hours. Workup of the reaction as described in Example 5 and purification of the products on silica gel (150 g), as described in Example 1, using a gradient of ethyl acetate in hexanes (20 to 60%) yielded three major fractions. Fraction 1(1.55 g) contained the faster isomer; fraction 2(0.90 g) contained a mixture of both isomers; fraction 3(1.53 g) contained the slower isomer. Overall yield of methyl 5-(cyano[2-(1-naphthyl)ethyl]methyl)-1-(2-deoxy-3,5 di-O-p-β-D-erythr o-pentofuranosyl) imidazole-4-carboxylate, 3.9 g, 42%. Fraction 1. 1 H-NMR: δ,8.17 (s, 1H, C(2)-H); 7.9-7.1 (m, 15H, aromatic); 6.38 (t, 1H, H-1'); 5.17 (t, 1H, methene); 3.72 (s, 3H, COOCH3). Fraction 3. δ,8.15 (s, 1H, H-2); 7.9-7.1 (m, 15H, aromatic);. 6.26 (t, 1H, H-1'); 4.82 (t, 1H, methene); 3.77 (s, 3H, COOCH3). Anal. Calcd for C40 H37 N3 O7 (671.75): C, 71.52; H, 5.55; N, 6.26. Found: C,71.76; H, 5.54; N, 6.02.

General Ammonolysis Method. 5-(Cyano[nonyl]methyl)-1-(2'deoxy-β-D-e ythro-pentofuranosyl) imidazole-4-carboxamide

The nucleoside methyl 5-(cyano[nonyl]methyl)-1-(2,'-deoxy-3,5-di-O-p-totuoyl-β-D-erythro-pe ntofuranosyl) imidazole-4-carboxylate (2.98 g, 4.6 mmol) was dissolved in anhydrous methanol (5 mL) and transferred to a stainless steel bomb. The solution was cooled to -78°C and then treated with anhydrous liquid ammonia (45 mL). The bomb was sealed and then heated to 100° C. in an oil bath for 21 hours. TLC (solvent B) exhibited products Rf=0.45, and toluamide Rf=0.85, indicating a complete removal of the toluoyl protecting groups. Ammonia was evaporated at room temperature and the amber gum which resulted was flash chromatographed on silica gel (80 g) using a gradient of methanol in ethyl acetate (5 to 10%). Fractions corresponding to the products were pooled and evaporated in vacuo to yield 5-(Cyano[nonyl]methyl)-1-(2'deoxy-β-D-erythro-pentofuranosyl) imidazole-4-carboxamide as a white foam, 1.2 g (63%). 1 H-NMR: δ,8.09. and 8.05 (s,s; 1H; H-2); 6.14 (t, 1H; H1'); 5.45 and 5.32(2 dd, 1H, methene); 2.10,1.80,1.40-1.05 and 0.82(4m, 19H; nonyl). Anal. Calcd. for C20H32N404 (392.50): C, 61.20; H, 8.22; N, 14.27. Found: C, 60.97; H, 8.24; N, 13.98.

Synthesis of 5-(Allyl[cyano]methyl)-1-(2'-deoxy-β-D-eythro-pentofuranosyl)-imidazo le-4 carboxamide

The nucleoside methyl 5-(allyl[cyano]methyl)-1-(2'deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pent ofuranosyl) imidazole-4-carboxylate (3.95 g, 7.08 mmol) was treated with liquid ammonia and heated to 100°C in a stainless steel bomb for 8 hours. The products of this reaction were worked up and purified on silica gel (80 g) as described in Example 1. The deprotected compound 5-(allyl[cyano]methyl)-1-(2'-deoxy-β-D-erythro-pentofuranosyl)-imidaz ole-4 carboxamide was isolated as a white foam, 1.3 g (58%). 1 H-NMR: δ,8.09 and 8.05 (s,s; 1 H; H-2); 6.15 (t, 1 H; H1'); 5.75 and 5.10 (2 m, 3 H; vinyl); 5.42 and 5.34 (t,t; 1 H; methene). Anal. Calcd for C14 H18 N4 O4 (306.33): C, 54.89; H, 5.92; N, 18.29. Found: C, 54.58; H, 5.92; N, 17.93.

Synthesis of 5-(Benzyl[cyano]methyl)-1-(2'-β-D-eythro-pentofuranosyl)-imidazole-4- carboxamide

The nucleoside methyl 5-(benzyl[cyano]methyl)-1-(2'-deoxy-3, 5di-o-p-toluoyl-β-D-erythro-pentofuranosyl) imidazole-4-carboxylate (3.0 g, 4.93 mmol) was treated with liquid ammonia and heated to 100°C in a stainless steel bomb for 6 hours. The products of this reaction were worked up and purified on silica gel (80 g) as described in Example 1. The deprotected compound 5-(benzyl[cyano]methyl)-1-(2'-β-D-erythro-pentofuranosyl)-imidazole-4 -carboxamide was isolated as a white foam, 1.0 g (59%). 1 H-NMR: δ,8.05 and 8.03 (s,s; 1H; H-2); 7.25 (m; 5H; phenyl); 6.17 and 6.07 (t,t; 1H; H1'); 5.50 and 5.42 (t,t; 1H; methines). Anal. Calcd for C18 H33 N3 O7 (357.39): C, 60.49; H, 5.92; N, 15.68. Found: C, 60.65; H, 5.69; N, 15.23.

Synthesis of 5-(Cyano[2-(1-naphthyl)ethyl]methyl)-1-(2deoxy-β-D-erythro-pentofuran osyl)-imidazole-4-carboxamide

The nucleoside methyl 5-(cyano[2-(1-naphthyl) ethyl]methyl)-1-(2-deoxy-3,5di-O-p-β-D-erythro-pentofuranosyl) imidazole-4-carboxylate (3.78 g, 5.76 mmol) was treated with liquid ammonia and then heated to 100°C in a stainless steel bomb for 20 hours. The products of this reaction were worked up and purified on silica gel (86 g) as described in Example 1. The deprotected compound 5-(cyano[2-(1-naphthyl) ethyl]methyl)-1-(2deoxy-β-D-erythro-pentofuranosyl)-imidazole-4-carbo xamide was isolated as a white foam, 1.2 g (49%). 1 H-NMR: δ,8.14 and 8.08 (s,s; 1H; H-2); 8.0-7.3 (m, 7H; naphthyl); 6.22 and 6.19 (t,t; 1H; H1'); 5.55 and 5.30 (t,t; 1H, methene). Anal. Calcd for C23 H24 N4 O4 (420.47): C, 65.70; H, 5.75; N, 13.32. Found: C, 65.78; H, 5.72; N, 13.05.

General Cyclization Method. 6-Amino-7-nonyl-1-(2-deoxy-β-D-ezythro-pentofuranosyl) imidazo[4,5-c]pyridin-4(5H)-one (2'-Deoxy-3-nonyl-3-deazaguanosine)

A solution of the nucleoside 5-(cyano[nonyl]methyl)-1-(2'deoxy-β-D-erythro-pentofuranosyl)imidazol e-4-carboxamide (250 mg) in 35 mL methanolic ammonia(saturated at -20°C) was heated to 95°C in a sealed vessel for 18 hours. The mixture was evaporated to afford a dark solid which was redissolved in hot ethanol. The solid which separated upon cooling was filtered and dried in vacuo for 18 hours to afford 2'-Deoxy-3-nonyl-3-deazaguanosine (180 mg, 72%) as an amorphous solid. MP 150°C dec. 1 H-NMR: δ,10.3 (bs, 1H, N-H); 7.90 (s, 1H, H-8); 6.08 (pseudo t, 1H, H-1', J=6.0 Hz); 5.15 (bs, 2H, NH2); 2.52, 1.30 and 1.82(3 m, 19H, nonyl). UV, .lambda. max,nm, (log ε): MeOH, 312(4.001), 278(4.040); pH 1, 322(3.687), 296 (3.845); pH 7, 308(2.811), 276(2.842); pH 12,290(3.978). Anal. Calcd for C20 H32 N4 O4 (392.50): C, 61.20; H, 8.22; N, 14.27. Found: C, 61.02; H, 8.20; N, 14.22.

Synthesis of 7-Allyl-6-amino-1-(2deoxy-β-D-erythro-pentofuranosyl) imidazole [4,5-c]pyridin-4-(5H)-one (2'-deoxy-3-allyl-3-deazaguanosine)

The nucleoside 5-(Allyl[cyano]methyl)-1-(2'-deoxy-β-D-erythro-pentofuranosyl)-imidaz ole-4 carboxamide (250 mg) was cyclized and purified according to the procedure described in Examples 13 and 1, respectively, to yield 7-Allyl-6-amino-1-(2deoxy-β-D-erythro-pentofuranosyl) imidazole [4,5-c]pyridin-4-(5H)-one (2'-deoxy-3-allyl-3-eazaguanosine) (190 mg, 76%) as an amorphous solid. MP.155°C dec. 1 H-NMR (200 mHz): δ,10.4 (bs, 1H, N-H); 7.98 (s, 1H, H-8); 6.05 (pseudo t, 1 H, H1', J=5.4 Hz); 5.92, 5.02 and 4.88 (3m, 3H, vinylic); 5.34 (s, 2H, NH2); 3.40 and 3.19 (2m, 2H, allylic). UV, .lambda. max,nm, (log ε): 310 (3.996), 276 (4.060); pH 1, 320 (3.891), 292, (4.064); pH 7, 306 (3.963), 274 (4.011); pH 12, 288 (4.160). Anal. Calcd for C14 H18 N4 O4 (306.32): C, 54.89; H, 5.92; N, 18.29. Found: C, 54.58; H, 92; N, 18.09.

Synthesis of 6-Amino-7-benzyl-1-(2-deoxy-β-D-erythro-pentofuranosyl) imidazo-[4,5c]pyridin 4(5H)-one (2'-deoxy-3-benzyl-3-deazaguanosine

The nucleoside 5-(Benzyl[cyano]methyl)-1-(2'-β-D-erythro-pentofuranosyl)-imidazole-4 -carboxamide (250 mg) was cyclized and purified by trituration with hot ethanol as described in Examples 13 to yield 2'-deoxy-3-benzyl-3-deazaguanosine (200 mg, 80%) as a white solid. MP 140°C dec. 1 H-NMR: δ 10.4 (bs, 1H, N-H); 7.84 (s, 1H, H-8); 7.3-7.1 (m, 5H, phenyl); 6.04 (pseudo t, 1H, H-1', J=6.3 Hz); 5.07 (bs, 2H, NH2); 4.07 and 3.88 (dd, 2H, benzylic, J=17 Hz). UV, .lambda. max,nm, (log ε): MeOH, 310 (3.950), 276 (4.002); pH 1, 320 (3.835), 294 (3.988); pH 7 308 (3.923), 274 (3.959); pH 12,310 (3.930). Anal. Calcd for C18 H20 N4 O4 (356.38): C, 60.66; H, 5.66; N, 15.72. Found: C, 60.15; H, 5.60; N, 15.51.

Synthesis of 6-Amino 7-(2-[1-naphthyl]ethyl)-1-(2deoxy-β-D-erythro-pentofuranosyl) imidazo[4,5-c]pyridin-4(5H)-one (2'-deoxy-3-(2-[1-naphthyl]ethyl)3-deazaguanosine

The nucleoside 5-(Cyano[2-(1-naphthyl) ethyl]methyl)-1-(2deoxy-β-D-erythro-pentofuranosyl)-imidazole-4-carbo xamide (250 mg) was cyclized and purified from hot ethanol as described in Example 13 to yield 6-Amino 7-(2-[1-naphthyl]ethyl)-1-(2deoxy-β-D-erythro-pentofuranosyl) imidazo[4,5-c]pyridin-4(5H)-one (2'-deoxy-3-(2-[1-naphthyl] ethyl)3-deazaguanosine (200 mg, 80%) as an off-white solid. MP 195° C. dec. 1 H-NMR: δ,10.5 (bs, 1H, N-H); 7.98 (s, 1H, H-8); 8.2-7.4 (m, 7H, naphthyl); 6.21 (pseudo t, 1H, H-1', J=6.9 Hz); 5.32 (bs, 2H, NH2); 3.3-2.8 (m, 4H, ethylene). UV .lambda. max,nm, (log ε): MeOH, 310(4.019), 289(4.180); pH 1,324(3.805), 290(4.114); pH 7,310 (3.611), 284(3.834); pH 12,286(4.185). Anal. Calcd for C23 H24 N4 O4 (420.47): C, 65.70; H, 5.75; N, 13.32. Found: C, 65.80; H, 5.72; N, 13.05.

General Heterocycle Alkylation Method. Methyl 5-(cyano[nonyl]methyl)-3-tetrahydropyranyl-imidazole-4-carboxylate

The cyanomethyl imidazole (5.0 g, 20 mmol) was alkylated using NaH (1.2 g, 60% in oil, washed with hexanes) and iodononane (7.6 g, 30 mmol) as described in Example 5, the General Procedure for the nucleosides. The products were purified by FC using a gradient of ethyl acetate in hexanes (20 to 50%). Evaporation of the fractions containing the products yielded methyl 5-(cyano[nonyl]methyl)-3-tetrahydro-pyranyl-imidazole-4-carboxylate (5.0 g, 67%) as a yellow foam. 1 H-NMR (200 mHz): δ,8.22 (s, 1H, H-2); 5.87 (m, 1 H, H-2' tetrahydropyranyl); 4.58 (m, 1H, methene); 3.85 (s, 3H, COOCH3); 1.30 and 0.88(2 m, 19H, nonyl). Anal. Calcd for C21 H33 N3 O3 (375.51): C, 67.17; H, 8.86; N, 11.19. Found: C, 67.47; H, 8.95; N, 11.21.

Synthesis of Methyl 5-(allyl[cyano]methyl)-3-tetrahydro-pyranyl-imidazole-4-carboxylate

The cyanomethyl imidazole (5.0 g, 20 mmol) was alkylated using NaH (1.2 g) and allyl bromide (3.6 g, 30 mmol) and purified according to Examples 5 and 1, respectively. White foam, 3.9 g, 67%. 1 H-NMR (200 mHz): δ,8.23 (s, 1H, H-2); 5.87 (m, 1H, H-2' tetrahydropyranyl); 5.77 and 5.15 (2m, 3H, vinylic); 5.68 (m, 1H, methene); 3.86 (s, 3H, COOCH3). Anal. Calcd for C15 H19 N3 O3 : C, 62.27; H, 6.62; N, 14.52. Found: C, 62.27; H, 6.62; N, 14.54.

Synthesis of Methyl 5-(benzyl[cyano]methyl)-3-tetrahydropyranyl-imidazole-4-carboxylate

The cyanomethyl imidazole (5.0 g, 20 mmol) was alkylated using NaH (1.2 g) and benzyl bromide (5.1 g, 30 mmol), as described in Example 5, and purified according to Example 1. White foam, 4.8 g, 71%. 1 H-NMR (200 mHz): δ,8.25 (s, 1H, H-2); 7.29 (m, 5H, phenyl); 6.83 (m, 1H, H-2' tetrahydropyranyl); 4.84 (m, 1H, methene); 3.84 and 3.80 (s,s; 3H, COOCH3); 3.19 (m, 2H, benzylic). Anal. Calcd. for C19 H21 N3 O3 (339.39): C, 67.24; H, 6.24; N, 12.38. Found: C, 67.21; H, 6.19; N, 12.02.

Synthesis of Methyl 5-(cyano[2-(1-naphthyl)ethyl]methyl)-3-tetrahydropyranyl -imidazole-4-carb oxylate

The cyanomethyl imidazole (5.0 g, 20 mmol) was alkylated using NaH (1.2 g) and 2-(1-naphthyl)ethyl bromide (7.1 g, 30 mmol, Frinton Labs, Piscataway, N.J.), as described in Example 5, and purified according to the procedure discussed in Example 1. White foam, 5.0 g, 62%. 1 H-NMR (200 mHz): δ,8.23 (s, 1H, H-2); 8.1-7.3 (m, 7H, naphthyl); 5.80 (m, 1H, H-2' tetrahydropyranyl); 4.58 (m, 1H, methene); 3.57 (s, 3H, COOCH3). Anal. Calcd for C24 H35 N3 O3 (403.48): C, 71.44; H, 6.24; N, 10.41. Found: 71.38; H, 6.16; N, 10.45.

General Nucleoside Cyclization Method. 6-Amino-7-nonyl-1,5-dihydroimidazo [4,5-c]pyridin-4-one Hydrochloride Salt

The compound methyl 5-(cyano[nonyl]methyl)-3-tetrahydropyranyl-imidazole-4-carboxylate (2.5 g, 6.7 mmol) was dissolved in 50 mL methanolic ammonia (saturated at -20°C) in a stainless steel bomb. The mixture was stirred at 75°C for 72 hours and then evaporated to dryness In vacuo. The resulting solid was co-evaporated with methanol (45 mL) and then immediately stirred with methanolic 1N HCl (50 mL) for 12 hours. After this time, the reaction mixture was evaporated to afford a light yellow gum. This gum was precipitated by repeated co-evaporation with methanol. Amorphous solid, 1.1 g, 53%. MP 225 darkens; 240°C dec. 1 H-NMR: δ,10.8 (bs, 1H, N-H); 8.72 (s, 1H, H-2); 5.80 (bs, 2H, NH2); 2.0, 0.9 and 0.4(3m, 19H, nonyl). UV, .lambda. max,nm, (log ε): MeOH, 310(3.935), 270(3.969). Anal. Calcd. for C15 H25 N4 OC1 (312.84): C, 57.59; H, 8.05; N, 17.90. Found: C, 57.53; H, 8.05; N, 17.87.

Synthesis of 7-Allyl-6-amino-1,5-dihydroimidazo [4,5-c] pyridin-4-one Hydrochloride Salt

The compound methyl 5-(allyl[cyano]methyl)-3-tetrahydropyranyl-imidazole-4-carboxylate (2.5 g, 8.6 mmol) was cyclized and isolated according to the procedure discussed in Example 21. Amorphous solid, 1.3 g, 67%. MP 210°C dec. 1 H-NMR: δ,11.0 (bs, 1 H, NH); 8.49 (s, 1H, H-2); 5.80 and 4.98(2m, 3H, vinylic); 5.80 (bs, 2H, NH2); 3.26 (m, 2H, allylic). UV .lambda. max,nm, (log ε): MeOH, 310(3.927), 268 (3.985). Anal. Calcd for C9 H11 N4 OC1 (226.66): C, 47.69; H, 4.89; N, 24.72. Found: C, 48.04; H, 5.02; N, 23.76.

Synthesis of 6-Amino-7-benzyl-1,5-dihydroimidazo[4,5c] pyridin-4-one Hydrochloride Salt

The compound methyl 5-(benzyl[cyano]methyl)-3-tetrahydropyranyl-imidazole -4-carboxylate (2.5 g, 7.4 mmol) was cyclized and isolated according to the procedure described in Example 21. Amorphous solid, 1.9 g, 92%. MP 196°C dec. 1 H-NMR: δ,11.5 (bs, 1 H, N-H); 9.20 (s, 1H, H-2); 7.17 (m, 5H, phenyl); 6.20 (bs, 2H, NH2); 3.95 (s, 2H, benzylic). UV, .lambda. max,nm, (log ε): MeOH, 310(3.958), 268(3.988). Anal. Calcd for C13 H13 N4 OC1 (276.72): C, 56.42; H, 4.73; N, 20.25. Found: C, 56.40; H, 4.71; N, 20.03.

Synthesis of 6-Amino-7-(2-[1-naphthyl]ethyl)-1,5-dihydro[4,5-c]pyridin-4-one Hydrochlor ide Salt

The compound methyl 5-(cyano[2-(1-naphthyl)ethyl]methyl)-3-tetrahydropyranyl-imidazole-4-carbo xylate (2.5 g, 6.2 mmol) was cyclized and isolated according to the procedure described in Example 21. Amorphous solid, 1.64 g, 78%. MP 260 darkens, 273 °C dec. 1 H-NMR: δ,11.4 (bs, 1H, N-H); 9.20 (s, 1H, H-2); 8.1-7.4 (m, 7H, naphthyl); 6.10 (bs, 2H, NH2); 3.15 and 3,02 (2 m, 4H, ethylene). UV, 80 max,nm, (log ε): MeOH, 308 (3.930), 272 (4.137). Anal. Calcd for C18 H17 N4 OC1 (340.81): C, 63.18; H, 4.95; N, 16.28. Found: C, 63.18; H, 4.95; H, 16.28.

Synthesis of 6-Amino-1,5-dihydro[4,5-c]pyridin-4-one (3-Deazaguanine) Hydrochloride Salt

The compound cyanomethyl imidazole (2.5 g, 10 mmol) was cyclized and the product isolated according to the procedure described in Example 21. This compound gave a satisfactory 1 H-NMR but proved unstable during purification and did not fit C,H,N analysis as a hydrochloride salt, Hartman, supra: 1 H-NMR: δ,10.5 (bs, 1H, N-H); 8.18 (s, 1H, H-2); 5.4 (bs, 2H, NH2); 4.6 (s, 1H, H-7).

Synthesis of Methyl 5-(cyano[nonyl]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pen tofuranosyl) imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl) -1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuranosyl) imidazole-4-carboxylate (5.7 g, 11 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.88 g, 60% in oil, washed with hexanes) at room temperature and under an atmosphere of argon. This suspension was stirred for 15 minutes and then treated with iodononane (7.5 mL, 37.4 mmol) using a syringe. The reaction mixture was stirred under these conditions for 6 hr; thin layer chromatography plates (ethyl acetate/hexanes, 3:2, v/v) showed the disappearance of starting material nucleoside and the appearance of two faster migrating products. The reaction was quenched with the addition of glacial acetic acid to pH 5 and then evaporated to dryness in vacuo to afford a yellow syrup. The syrup was dissolved in dichloromethane (150 mL) and the solution was washed with cold 0.1N HCl, water, and then dried over magnesium sulfate. Filtration of the desiccant and evaporation of the solvent afforded a yellow gum which was flash-chromatographed on silica gel (120 g) using ethyl acetate-hexanes (1:4 then 1:1). Fractions corresponding to the alkylated products were pooled and evaporated to yield Methyl 5-(cyano[nonyl]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pen tofuranosyl) imidazole-4-carboxylate. as a yellowish foam, 2.98 g (47%). 1H-NMR (Me2 SO-d6): δ, 8.18 and 8.15 (s,s; C-2 H, 1 H); 6.48 and 6.37 (t,t; H-1', 1 H); 1.18 and 0.90 (2 m, nonyl, 19 H).

Synthesis of Methyl 5-(allyl[cyano]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pen tofuranosyl)imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate (5.0 g, 9.7 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.46 g, 11.6 mmol) and then allyl bromide (2.5 mL, 29 mmol) in the manner described for in Example 29. The reaction was worked up and chromatographed on silica gel (75 g) using the aforementioned solvent system to afford 84 as a yellowish foam, 3.66 g (68%). 1H-NMR (Me2 SO-d6):δ,8.15 and 8.13 (s,s; C-2 H, 1 H); 6.38 (m, H-1', 1 H); 5.75 and 5.08 (2 m, vinyl, 3 H).

Synthesis of Methyl 5-(benzyl[cyano]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pe ntofuranosyl) imidazole-4-carboxylate

A solution of methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole-4-carboxylate (5.0 g, 9.6 mmol) in anhydrous acetonitrile (75 mL) was treated with sodium hydride (0.46 g, 11 mmol) under argon and stirred at room temperature for 15 minutes. The mixture was cooled to 4°C in an ice bath and a solution of benzyl bromide (1.26 mL, 10.6 mmol) in acetonitrile (15 mL) was added dropwise over 70 min. The ice bath was removed and the reaction further stirred at room temperature for 2.5 hours. Workup of the reaction and purification of the products on silica gel (100 g) afforded methyl 5-(benzyl[cyano]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pe ntofuranosyl) imidazole-4-carboxylate. as a white foam, 3.4 g (58%). 1H-NMR (Me2 SO-d6): δ,8.10 and 8.05 (s,s; C-2 H, 1 H); 7.30-7.10 (m, phenyl, 5 H); 5.33 and 6.01 (t,t; H-1', 1 H).

Synthesis of 5-(Cyano[nonyl]methyl)-1-(2'-deoxy-β-D-erythropento-furanosyl) imidazole-4-carboxamide

The nonyl-imidazole nucleoside, methyl 5-(cyano[nonyl]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pen tofuranosyl) imidazole-4-carboxylate, (2.98 g, 4.6 mmol) was dissolved in anhydrous methanol (5 mL) and trans-ferred to a stainless steel bomb. The solution was cooled in a dry-ice /isopropanol bath and then treated with anhydrous liquid ammonia (45 mL). The bomb was sealed and then heated to 100°C in an oil bath for 21 hours. TLC (ethyl acetate/methanol, 4:1, v/v) indicated a complete removal of the toluoyl protecting groups. Excess ammonia was evaporated at room temperature and the amber gum which resulted was flash chromatographed on silica gel (80 g) using ethyl acetate/methanol (95:5 then 9:1). Fractions corresponding to deblocked products were pooled and evaporated in vacuo to yield 5-(cyano[nonyl]methyl)-1-(2'-deoxy-β-D-erythropento-furanosyl) imidazole-4-carboxamide as a white foam, 1.14 g (63%). 1H-NMR (Me2 SO-d6):δ,8.09 and 8.05 (s,s; C-2 H, 1 H); 6.14 (t, H-1', 1H); 1.40-1.05 and 0.95 (2 m, nonyl, 19 H).

Synthesis of 5-(Allyl[cyano]methyl)-1-(2'-deozy-β-D-erythropento-furanosyl) imidazole-4-carboxamide

The allyl-imidazole nucleoside methyl 5-(allyl [cyano]methyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofurano syl) imidazole-4-carboxylate (3.95 g, 7.08 mmol) was treated with liquid ammonia in a stainless steel bomb and heated to 100°C for 8 hours. The products of this reaction were worked up and purified on silica gel (80 g) in a manner analogous to that for compound synthesized according to Example 32, 5-(Cyano[nonyl]methyl)-1-(2'-deoxy-β-D-erythropento-furanosyl) imidazole-4-carboxamide. The de-blocked compound synthesized according to Example 33, 5-(Allyl[cyano]methyl)-1-(2'-deoxy-β-D-erythropento-furanosyl) imidazole-4-carboxamide, was isolated as a white foam, 1.29 g (58%). 1H-NMR (Me2 SO-d6):δ,8.09 and 8.07 (s,s; C-2 H, 1 H); 6.15 (t, H-1', 1 H); 5.75 and 5.10 (2 m, vinyl, 3 H).

Synthesis of 5-(Benzyl[cyano]methyl)-1-(2'-deoxy-β-D-erythropento-furanosyl) imidazole-4-carboxamide

The benzyl-imidazole nucleoside synthesized according to Example 31 (3.0 g, 4.93 mmol) was treated with liquid ammonia in a stainless steel bomb and heated to 100°C for 6 hours. The products of this reaction were worked up and purified on silica gel (80 g) in a manner analogous to that for compound synthesized according to Example 32. The deblocked compound synthesized according to Example 34 was isolated as a white foam, 1.03 g (59%). 1H-NMR (Me2 SO-d6): δ,8.03 and 8.04 (s,s; C-2 H, 1 H); 7.25 (m, phenyl, 5 H); 6.17 and 6.07 (t,t; H-1', 1 H).

Synthesis of 5-(cyanomethyl)-1-(2'-deoxy-5'-O-dimethoxytrityl-β-D-erythropentofura nosyl) imidazole-4-carboxamide

The nucleoside, methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole -4-carboxamide, (1.95 g, 7.3 mmol) was dried by co-evaporation with pyridine (30 mL). The gum which resulted was dissolved in anhydrous pyridine under argon and then treated with dimethoxytrityl chloride (2.90 g, 12.4 mmol). The mixture was stirred at room temperature for 2 hours after which TLC (ethyl acetate:methanol, 19:1, v/v) indicated complete conversion of starting material. Tritylated products were visualized as orange spots using H2 SO4 fumes. The reaction was quenched with the addition of methanol (2 mL) followed by stirring for 15 min. The mixture was evaporated in vacuo to afford a thick orange syrup which was co-evaporated with toluene (3×25 mL). The syrup was flash chromatographed on silica gel (100 g) using a stepwise gradient of methanol in 1% Et3 N/CH2 Cl2 (0 to 5% methanol). The appropriate fractions were pooled and evaporated to yield 5-(Cyanomethyl)-1-(2'-deoxy-5'-O-dimethoxytrityl-β-D-erythropentofura nosyl) imidazole-4-carboxamide as a white foam, 3.64 g (87%). 1H-NMR (Me2 SO-d6): δ,7.96 (s, C-2 H, 1 H); 6.85-7.35 (m, DMT, 13 H); 6.13 (t, H-1', 1 H).

Synthesis of 5-(Cyano[nonyl]methyl)-1-(2'-deoxy-5'-O-dimethoxytrityl-β-D-erythrope ntofuranosyl) imidazole-4-carboxamide

The nucleoside synthesized according to Example 32 (1.20 g, 3.1 mmol) was thoroughly dried by co-evaporation with anhydrous pyridine (30 mL). The syrup which resulted was redissolved in anhydrous pyridine under argon and treated with dimethoxytrityl chloride (1.0 g, 3.1 mMol). The reaction mixture was stirred at room temperature for 3.5 hr, after which time TLC (ethyl acetate) indicated complete conversion of the starting material. The reaction mixture was treated with 2 mL of anhydrous methanol, stirred for 15 minutes and then evaporated in vacuo to afford a bright orange syrup. This syrup was co-evaporated with toluene (2×50 mL) and then flash chromatographed on silica (80 g) using a step-wise gradient of methanol in dichloromethane/1% triethylamine (0 to 3% methanol). The appropriate fractions were pooled and evaporated in vacuo to yield the dimethoxy-tritylated compound 5-(Cyano[nonyl]methyl)-1-(2'-deoxy-5'-O-dimethoxytrityl-β-D-erythrope ntofuranosyl) imidazole-4-carboxamide as a white foam, 1.46 g (69%). 1H-NMR (Me2 SO-d6): δ,7.98 and 7.93 (s,s; C-2 H, 1 H); 7.30 and 6.92 (2 m,DMT, 13 H); 6.21 (t, H-1', 1 H); 1.20 and 0.92 (2 m, nonyl, 19 H).

Synthesis of 5-(Allyl[cyano]methyl)-1-(2'-deoxy-5'-O-dimethoxytrityl-β-D-erythrope ntofuranosyl) imidazole-4-carboxamide

The nucleoside synthesized according to Example 33 (1.25 g, 4.08 mmol) was dried by co-evaporation with pyridine and then redissolved in anhydrous pyridine (50 mL) and treated with dimethoxy-trityl chloride (1.38 g, 4.08 mmol) under an atmosphere of argon. The reaction was stirred for 2.5 hours and then worked up and products isolated by flash chromatography on silica gel (90 g) in a manner analogous to compound synthesized according to Example 35. The appropriate fractions were pooled and evaporated in vacuo to yield dimethoxytritylated compound 37 as a white foam, 1.86 g (75%). 1H-NMR (Me2 SO-d6):δ,7.98 and 7.95 (s,s; C-2 H, 1 H); 7.25 and 6.93 (2 m, DMT, 13 H); 6.21 (m, H-1', 1 H); 5.78 and 5.10 (2 m, vinyl, 3 H).

Synthesis of 5-(Benzyl[cyano]methyl)-1-(2'-deoxy-5'-O-dimethoxy-trityl-β-D-erythro pentofuranosyl) imidazole-4-carboxamide

The nucleoside synthesized according to Example 34, 930 mg (2.6 mmol) was dried by co-evaporation with pyridine and then redissolved in anhydrous pyridine (50 mL) and treated with dimethoxy-trityl chloride (884 mg, 2.6 mmol) under an atmosphere of argon. The reaction was stirred for 4 hours and then worked up and the products isolated by flash chromatography on silica gel (80 g) in a manner analogous to the compound synthesized according to Example 35. The appropriate fractions were pooled and evaporated in vacuo to yield 1.50 g of 5-(Benzyl[cyano]methyl)-1-(2'-deoxy-5'-O-dimethoxy-trityl-β-D-erythro pentofuranosyl) imidazole-4-carboxamide as a pinkish foam, (87%). 1H-NMR (Me2 SO-d6):δ,7.70 (s, C-2 H, 1 H); 7.40 and 6.70 (2m; DMT, phenyl; 18 H); 6.10 (t, H-1', 1 H).

Synthesis of 5-(Cyanomethyl)-1-(5'-O-dimethoxytrityl-3'-O-[2-cyano-ethyl-N,N-diisopropy l]phosphoramidite-2'-deoxy-β-D-erythropentofuranosyl) imidazole-4-carboxamide

The tritylated nucleoside synthesized according to Example 35 (1.82 g, 3.2 mmol) was dissolved in anhydrous tetrahydrofuran (50 mL) under argon and then treated with diisopropyl ethylamine (1 mL). The solution was cooled to 4°C in an ice bath and then treated with 2-cyanoethyl-N,N-diisopropyl phosphorochloridate (1.2 g, 5.12 mmol) in one portion. The ice bath was removed and the reaction mixture was stirred at room temperature for 3 hours. At the end of this time, TLC (CH2 Cl2 /1% MeOH/1% Et3 N) indicated complete conversion of starting material. Reaction products were visualized using H2 SO4 fumes. The reaction mixture was evaporated in vacuo to afford a thick syrup which was immediately redissolved in dichloromethane (100 mL) and washed with cold, saturated sodium bicarbonate (2×50 mL) and brine (50 mL). The organic layer was dried over magnesium sulfate, filtered and evaporated to yield a yellowish foam (2.8 g). This foam was flash chromatographed on silica gel (75 g) using a stepwise gradient of ethyl acetate/hexanes (1:4 to 1:1) containing 1% Et3 N. The appropriate fractions were pooled and evaporated to yield 5-(Cyanomethyl)-1-(5'-O-dimethoxytrityl-3'-O-[2-cyano-ethyl-N, N-diisopropyl]phosphoramidite-2'-deoxy-β-D-erythropentofuranosyl) imidazole-4-carboxamide. as a white foam, 1.65 g (59%). An aliquot of this material was precipitated by dissolving the foam in a small volume of dichloromethane and adding it to a large proportion (1:50) of hexanes. 1H-NMR (CD3 CN): δ,7.78 and 7.72 (s,s; C-2 H, 1H); 7.25 and 6.95 (m, DMT, 13 H); 6.12 (t, H-1', 1H). 31P-NMR (CD3 CN): δ, 150.02, 149.98.

Synthesis of 5-(Cyano[nonyl]methyl)-1-(5'-O-dimethoxytrityl-3'-[2-cyanoethyl-N,N-diisop ropyl]phosphoramidite-2'-deoxy-β-D-erythropentofuranosyl) imidazole-4-carboxamide

The tritylated nucleoside synthesized according to Example 36, 1.46 g (2.1 mmol) was dissolved in anhydrous tetrahydrofuran (50 mL) under argon and then treated with diisopropyl ethylamine (1.5 mL). This mixture was cooled to 4°C in an ice bath and then treated with 2-cyanoethyl-N,N-diisopropylamino phosphorochloridite (488 mg, 2.1 mmol) in one portion. The ice bath was removed and the reaction was further stirred at room temperature for a total of 3 hours. Additional phosphorochloridite (48 mg) was added at the end of the first and second hours of reaction. At the end of this time, TLC (CH2 Cl2 /3% MeOH/1% Et3 N) indicated complete conversion of starting material. The reaction mixture was worked up as described for the compound synthesized according to Example 39 and the products purified on silica gel (80 g) using a stepwise gradient of ethyl acetate/hexanes (2:3 to 3:2) containing 1% Et3 N. The appropriate fractions were pooled and evaporated to afford 5-(Cyano[nonyl]methyl)-1-(5'-O-dimethoxytrityl-31-0-[2-cyanoethyl-N, N-diisopropyl]phosphoramidite-2'-deoxy-β-D-erythropentofuranosyl) imidazole-4-carboxamide as a colorless foam, 1.37 g (73%). An aliquot of this material was precipitated using a procedure described for the compound synthesized according to Example 39. 1H-NMR (CD3 CN): δ, 7.75 (s, C-2 H); 7.25 and 6.85 (2 m, DMT, 13 H), 6.25 and 6.20 (2 m, H-1', 1 H). 31 P-NMR (CD3 CN): δ, 150.1, 150.0 and 149.9.

Synthesis of 5-(Ally[cyano]methyl)-1-(5'-O-dimethoxytrityl-3'-O-[2-cyanoethyl-N,N-diiso propyl]phosphoramidite-2'-deoxy-β-D-erythro-pentofuranosyl) imidazole-4-carboxamide

The tritylated nucleoside synthesized according to Example 37, 1.86 g (3.1 mmol) was dissolved in anhydrous THF (50 mL) and then treated with diisopropyl ethylamine (1.5 mL) and 2-cyanoethyl N,N-diisopropylamino phosphochloridite (705 mg, 3.1 mmol) in a manner analogous to the compound synthesized according to Example 40. The mixture was stirred at room temperature for a total of 3 hours. Additional phosphochloridite (140 mg) was added at the end of 2 hour reaction time. The reaction was worked up and the products isolated by flash chromatography on silica gel (80 g) using a stepwise gradient of ethyl acetate/hexanes (2:3 to 4:1, v/v). The appropriate fractions were pooled and evaporated to yield 2.18 g (88%) of 41 as a white solid foam. An aliquot of this material was precipitated using a procedure described for compound synthesized according to Example 39. 1H-NMR (CD3 CN): δ, 7.77 and 7.75 (s,s; C-2 H, 1 H); 7.45 and 6.85 (2m, DMT, 13 H); 6.25 and 6.20 (t,t; H-1', 1 H). 31P-NMR (CD3 CN): δ, 150.05, 149.98 and 149.85.

Synthesis of 5-(Benzyl[cyano]methyl)-1-(5'-O-dimethoxytrityl-3'-O-[2-cyanoethyl-N,N-dii sopropyl]phosphoramidite-2'-deoxy-β-D-erythro-pentofuranosyl) imidazole-4-carboxamide

The tritylated nucleoside synthesized according to Example 38, 1.50 g (2.3 mmol) was dissolved in anhydrous THF (50 mL) and then treated with diisopropyl ethylamine (1.5 mL) and 2-cyanoethyl-N,N-di-isopropylamino phosphochloridite (539 mg, 2.3 mmol) in a manner analogous to the compound synthesized according to Example 39. The mixture was stirred at room temperature for a total of 3 hours. Additional phosphorochloridite (110 mg) was added at the end of 2 hours reaction time. The reaction was worked up and the products isolated by flash chromatography on silica gel (40 g) using a stepwise gradient of ethyl acetate/hexanes (1:4 to 3:2, v/v). The appropriate fractions were pooled and evaporated to yield 5-(Benzyl[cyano]methyl)-1-(5'-O-dimethoxytrityl-3'-O-[2-cyanoethyl-N,N-dii sopropyl]phosphoramidite-2'-deoxy-β-D-erythro-pentofuranosyl) imidazole-4-carboxamide., 1.03 g (55%) as a colorless foam. An aliquot of this material was precipitated using a procedure described for the compound synthesized according to Example 39. 1H-NMR (CD3 CN): δ, 7.72 (s,s; C-2 H; 1 H); 7.30 and 6.80 (2 m; DMT, benzyl; 18 H); 6.10 (m, H-1', 1 H). 31 P-NMR (CD3 CN): δ, 150.00, 149.90 and 149.85.

Synthesis of Oligomers

Oligomers incorporating the nucleotides of Examples 39, 40, 41 and 42 were made using standard phorphoramidite chemistries on an Applied Biosystems 380B synthesizer. Average coupling efficiencies were 94% using a method which left the 5'-dimethoxytrityl group of each oligonucleotide on. After cleavage from the solid support, the oligomers were treated with excess concentrated ammonium hydroxide and then heated in a sealed vessel at 55°C for a minimum of 15 hours. This procedure removed all protecting groups from A, G and C bases and caused cyclization of the 5-cyano[alkyl]methyl imidazole to the 3-deaza-3-alkyl (or aryl) heterocycles. For each nucleotide, verification of this cyclization was made from the analysis of a 1H-NMR spectrum of a TG3C trimer containing a single 3-deaza-3-substituted 2'-deoxyguanosine base. Purification of these trityl-on oligomers was performed using a C-4 Waters Prepak cartridge (10 cm) using a gradient elution of acetonitrile in 50 mM triethyl ammonium acetate (pH 7.0) (4 to 48t over 60 min). The trityl group was removed by a treatment with 15% acetic acid and the oligomers were then precipitated from 70% ethanol.

Synthesis of 5-(Cyano[propylphthalimide]methyl)-1-(5'-O-dimethoxy-trityl-3'-O'[2-cyanoe thyl-N,N-diisopropyl]phosphoramidite-2'-deoxy-β-D-erythro-pentofuranos yl-imidazole-4-carboxamide

In a manner as per Example 29, the compound methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole -4-carboxylate can be treated with N-(3-bromopropyl)phthalimide instead of iodononane. After work-up as per Example 29 the product can be treated as per Examples 35 and 39 to yield the 5-(Cyano [propylphthalimide]methyl)-1-(5'-O-dimethoxy-trityl-3'-O '[2-cyanoethyl-N,N-diisopropyl]phosphoramidite-2'-deoxy-β-D-erythro-p entofuranosyl-imidazole-4-carboxamide. In addition to ring closure upon cleavage from the oligonucleotide synthesizer solid support, the phthalimide is also cleaved to amino as per other above examples. Further the alkylamino product can be chain extended to a polyalkylamine.

Synthesis of 5-(Cyano[imidizo-1-yl(propyl)]methyl)-1-(5'-O-dimethoxy-trityl-3'-O'[2-cya noethyl-N,N-diisopropyl]phosphor-amidite-2'-deoxy-β-D-erythro-pentofur anosyl-imidazole-4-carboxamide

In a manner as per Example 29, the compound methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole -4-carboxylate can be treated with 1-(3-bromopropyl)imidazole instead of iodononane. After work-up as per Example 29 the product can be treated as per Examples 35 and 39 to yield 5-(Cyano[imidizo-1-yl (propyl)]methyl)-1-(5'-O-dimethoxy-trityl-3'-O'[2-cyanoethyl-N,N-diisoprop yl]phosphoramidite-2'-deoxy-β-D-erythro-pentofuranosyl-imidazole-4-car boxamide.

Synthesis of 5-(Cyano[anthracen-2-yl(propyl)]methyl)-1-(5'-O-di-methoxy-trityl-3'-O'[2- cyanoethyl-N,N-diisopropyl]phosphoramidite-2'-deozy-β-D-erythro-pentof uranosyl-imidazole-4-carboxamide

In a manner as per Example 29, the compound methyl 5-(cyanomethyl)-1-(2'-deoxy-3,5-di-O-p-toluoyl-β-D-erythro-pentofuran osyl) imidazole -4-carboxylate can be treated with 2-(3-bromopropyl)anthracene instead of iodononane. After work-up as per Example 29 the product can be treated as per Examples 35 and 39 to yield 5-(Cyano[anthracen-2-yl (propyl)]methyl)-1-(5'-O-di-methoxy-trityl-3'-O'(2-cyanoethyl-N,N-diisopro pyl) phosphoramidite-2'-deoxy-β-D-erythro-pentofuranosyl-imidazole-4-carbo xamide.

Acevedo, Oscar L., Andrews, Robert S., Cook, P. Dan

Patent Priority Assignee Title
10006027, Mar 19 2014 University of Utah Research Foundation Methods for modulating Ataxin 2 expression
10017764, Feb 08 2011 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising bicyclic nucleotides and uses thereof
10023861, Aug 29 2011 Ionis Pharmaceuticals, Inc Oligomer-conjugate complexes and their use
10036019, Mar 17 2014 IONIS PHARMACEUTICALS, INC. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
10077236, Jul 15 2013 The Regents of the University of California Azacyclic constrained analogs of FTY720
10098958, Jan 08 2009 Northwestern University Delivery of oligonucleotide functionalized nanoparticles
10144933, Jan 15 2014 WAVE LIFE SCIENCES JAPAN, INC Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
10149905, Jan 15 2014 WAVE LIFE SCIENCES JAPAN, INC Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
10160969, Jan 16 2014 WAVE LIFE SCIENCES LTD Chiral design
10167309, Jul 13 2012 WAVE LIFE SCIENCES LTD Asymmetric auxiliary group
10182988, Dec 03 2013 AURASENSE THERAPEUTICS, INC ; EXICURE, INC Liposomal particles, methods of making same and uses thereof
10202599, Aug 11 2011 Ionis Pharmaceuticals, Inc Selective antisense compounds and uses thereof
10214745, Mar 15 2012 University of Florida Research Foundation; University of Florida Board of Trustees Treatment of brain derived neurotrophic factor (BDNF) related diseases by inhibition of natural antisense transcript to BDNF
10221416, Apr 24 2014 IONIS PHARMACEUTICALS, INC. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
10266825, Nov 05 2002 IONIS PHARMACEUTICALS, INC. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
10273474, Mar 14 2013 Biogen MA Inc Methods for modulating Tau expression for reducing seizure and modifying a neurodegenerative syndrome
10273483, Apr 08 2015 The University of Chicago; Northwestern University Compositions and methods for correcting limb girdle muscular dystrophy type 2C using exon skipping
10280192, Jul 19 2011 WAVE LIFE SCIENCES LTD. Methods for the synthesis of functionalized nucleic acids
10280423, May 01 2014 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating complement factor B expression
10287584, Nov 12 2014 IONIS PHARMACEUTICALS, INC.; The Board of Regents of the University of Texas System Compounds and methods for the modulation of COMP
10301622, Nov 04 2013 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (SNA)
10307434, Jul 06 2009 WAVE LIFE SCIENCES LTD. Nucleic acid prodrugs and methods of use thereof
10308934, Mar 19 2014 IONIS PHARMACEUTICALS, INC. Compositions for modulating Ataxin 2 expression
10322173, Jan 15 2014 WAVE LIFE SCIENCES JAPAN, INC Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
10329318, Dec 02 2008 WAVE LIFE SCIENCES LTD Method for the synthesis of phosphorus atom modified nucleic acids
10335368, Jun 08 2012 The Regents of the University of Michigan Ultrasound-triggerable agents for tissue engineering
10370656, Jun 08 2006 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
10370661, Jun 14 2005 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
10385341, Apr 01 2014 Biogen MA Inc Compositions for modulating SOD-1 expression
10385388, Dec 06 2013 Integrated DNA Technologies, Inc Cleavable competitor polynucleotides
10391116, Nov 24 2008 Northwestern University Polyvalent RNA-nanoparticle compositions
10398784, Sep 14 2011 NORTHWESTERN UNIVERITY Nanoconjugates able to cross the blood-brain barrier
10407678, Apr 16 2015 IONIS PHARMACEUTICALS, INC. Compositions for modulating expression of C9ORF72 antisense transcript
10407680, Sep 29 2016 Biogen MA Inc Compounds and methods for reducing Tau expression
10421821, Oct 30 2015 Genentech, Inc. Anti-HtrA1 antibodies and methods of use thereof
10421822, Oct 30 2015 Genetech, Inc. Anti-HtrA1 antibodies and methods of use thereof
10428019, Sep 24 2010 WAVE LIFE SCIENCES LTD Chiral auxiliaries
10435430, Jul 31 2013 The Board of Regents of the University of Texas System Methods and compounds useful in conditions related to repeat expansion
10472627, Dec 28 2005 University of Florida Board of Trustees; University of Florida Research Foundation Natural antisense and non-coding RNA transcripts as drug targets
10480030, Oct 27 2009 Integrated DNA Technologies, Inc Polynucleotide primers and probes
10513706, Apr 09 2014 The Scripps Research Institute Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
10517889, Sep 08 2017 Ionis Pharmaceuticals, Inc Modulators of SMAD7 expression
10533175, Sep 25 2015 IONIS PHARMACEUTICALS, INC.; Academisch Ziekenhuis Leiden Compositions and methods for modulating Ataxin 3 expression
10533178, Mar 19 2014 L&L Candle Company LLC Methods for modulating Ataxin 2 expression
10538763, Jan 16 2015 FRED HUTCHINSON CANCER CENTER Compounds and methods for modulation of DUX4
10557137, Nov 06 2015 IONIS PHARMACEUTICALS, INC. Modulating apolipoprotein (a) expression
10570169, May 22 2014 IONIS PHARMACEUTICALS, INC. Conjugated antisense compounds and their use
10577607, Mar 16 2016 IONIS PHARMACEUTICALS, INC. Modulation of DYRK1B expression
10590413, Jul 13 2012 WAVE LIFE SCIENCES LTD. Chiral control
10610571, Aug 03 2017 SYNTHORX, INC Cytokine conjugates for the treatment of proliferative and infectious diseases
10626138, Aug 08 2013 The Scripps Research Institute Method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
10669546, Apr 01 2014 BIOGEN MA INC. Compositions for modulating SOD-1 expression
10676738, Apr 28 2010 Ionis Pharmaceuticals, Inc 5′ modified nucleosides and oligomeric compounds prepared therefrom
10683499, May 01 2013 Glaxo Group Limited; Ionis Pharmaceuticals, Inc Compositions and methods for modulating TTR expression
10696719, Jun 24 2016 The Scripps Research Institute Nucleoside triphosphate transporter and uses thereof
10696720, Jun 24 2016 The Scripps Research Institute Nucleoside triphosphate transporter and uses thereof
10709812, Sep 29 2015 The Regents of the University of Michigan Biodegradable hydrogel for tissue expansion
10731166, Mar 18 2016 CARIS SCIENCE, INC Oligonucleotide probes and uses thereof
10772906, Oct 15 2008 IONIS PHARMACEUTICALS, INC. Modulation of Factor 11 expression
10792251, Dec 03 2013 Northwestern University; Exicure, Inc. Liposomal particles, methods of making same and uses thereof
10793855, Jan 06 2015 IONIS PHARMACEUTICALS, INC. Compositions for modulating expression of C9ORF72 antisense transcript
10793856, Jul 19 2013 Biogen MA Inc Compositions for modulating Tau expression
10793862, May 01 2014 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating growth hormone receptor expression
10801029, Apr 08 2015 The University of Chicago; Northwestern University Compositions and methods for correcting limb girdle muscular dystrophy type 2C using exon skipping
10813934, Feb 02 2011 The Trustees of Princeton University Sirtuin modulators as inhibitors of cytomegalovirus
10815480, Nov 17 2010 IONIS PHARMACEUTICALS, INC. Modulation of alpha synuclein expression
10865414, Jan 15 2018 Ionis Pharmaceuticals, Inc Modulators of DNM2 expression
10875884, May 01 2014 ISIS Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
10883104, May 01 2013 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating apolipoprotein (a) expression
10907160, Jan 05 2016 IONIS PHARMACEUTICALS, INC. Methods for reducing LRRK2 expression
10913951, Oct 31 2018 University of Pittsburgh - Of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
10941176, Jul 28 2015 CARIS SCIENCE, INC Therapeutic oligonucleotides
10954519, Aug 09 2013 lonis Pharmaceuticals, Inc. Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
10961271, Mar 16 2016 IONIS PHARMACEUTICALS, INC. Methods of modulating KEAP1
10968453, Apr 01 2014 BIOGEN MA INC. Compositions for modulating SOD-1 expression
10995068, Sep 24 2015 The Regents of the University of California; The Université de Montréal Synthetic sphingolipid-like molecules, drugs, methods of their synthesis and methods of treatment
11001622, Nov 19 2015 THE BRIGHAM AND WOMEN S HOSPITAL, INC Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
11021710, May 09 2018 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing FXI expression
11033570, Dec 02 2016 Cold Spring Harbor Laboratory; IONIS PHARMACEUTICALS, INC. Modulation of Lnc05 expression
11053498, Sep 29 2016 Biogen MA Inc Compounds and methods for reducing Tau expression
11058709, Dec 04 2015 IONIS PHARMACEUTICALS, INC.; Cold Spring Harbor Laboratory Methods of treating breast cancer
11066668, Jan 11 2012 Cold Spring Harbor Laboratory Compositions and methods for modulation of IKBKAP splicing
11077195, Feb 06 2019 SYNTHORX, INC. IL-2 conjugates and methods of use thereof
11078486, Jul 25 2018 Ionis Pharmaceuticals, Inc Compounds and methods for reducing ATXN2 expression
11083799, Mar 16 2016 CRISPR THERAPEUTICS AG Materials and methods for treatment of hereditary haemochromatosis
11084844, Apr 28 2010 IONIS PHARMACEUTICALS, INC. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
11091765, Jun 29 2015 Caris Science, Inc. Therapeutic oligonucleotides
11111494, Mar 19 2014 IONIS PHARMACEUTICALS, INC. Compositions for modulating Ataxin 2 expression
11129844, Mar 03 2015 IONIS PHARMACEUTICALS, INC.; Baylor College of Medicine Compositions and methods for modulating MECP2 expression
11136577, Mar 09 2016 IONIS PHARMACEUTICALS, INC. Methods and compositions for inhibiting PMP22 expression
11142800, Oct 07 2010 The General Hospital Corporation Biomarkers of cancer
11149264, Feb 12 2018 IONIS PHARMACEUTICALS, INC. Modified compounds and uses thereof
11155796, Feb 09 2015 Duke University Compositions and methods for epigenome editing
11155815, Mar 14 2013 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating Tau expression
11162096, Oct 14 2013 Ionis Pharmaceuticals, Inc; The Regents of the University of California; Ludwig Institute for Cancer Methods for modulating expression of C9ORF72 antisense transcript
11174469, Jun 29 2016 CRISPR THERAPEUTICS AG Materials and methods for treatment of Amyotrophic Lateral Sclerosis (ALS) and other related disorders
11180756, Mar 09 2017 Ionis Pharmaceuticals Morpholino modified oligomeric compounds
11193128, Oct 14 2013 IONIS PHARMACEUTICALS, INC. Compositions for modulating expression of C9ORF72 antisense transcript
11197884, Aug 18 2017 IONIS PHARMACEUTICALS, INC. Modulation of the notch signaling pathway for treatment of respiratory disorders
11208650, Nov 15 2018 Ionis Pharmaceuticals, Inc Modulators of IRF5 expression
11213593, Nov 21 2014 Northwestern University Sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
11214803, Jan 31 2019 Ionis Pharmaceuticals, Inc Modulators of YAP1 expression
11230712, Nov 09 2017 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing SNCA expression
11236335, Oct 12 2012 IONIS PHARMACEUTICALS, INC. Selective antisense compounds and uses thereof
11236339, Jun 17 2016 IONIS PHARMACEUTICALS, INC.; The Hospital for Sick Children Modulation of GYS1 expression
11241451, Mar 02 2018 IONIS PHARMACEUTICALS, INC. Modulators of IRF4 expression
11253601, Jul 11 2016 TRANSLATE BIO MA, INC Nucleic acid conjugates and uses thereof
11261446, Mar 29 2019 IONIS PHARMACEUTICALS, INC. Compounds and methods for modulating UBE3A-ATS
11268094, Apr 28 2010 Ionis Pharmaceuticals, Inc 5′ modified nucleosides and oligomeric compounds prepared therefrom
11279932, Feb 27 2019 Ionis Pharmaceuticals, Inc Modulators of MALAT1 expression
11293017, May 25 2016 CARIS SCIENCE, INC Oligonucleotide probes and uses thereof
11293025, Sep 25 2015 IONIS PHARMACEUTICALS, INC.; Academisch Ziekenhuis Leiden Compositions and methods for modulating Ataxin 3 expression
11299736, May 01 2013 IONIS PHARMACEUTICALS, INC. Conjugated antisense compounds and their use
11299737, Feb 28 2020 IONIS PHARMACEUTICALS, INC. Compounds and methods for modulating SMN2
11312964, May 01 2014 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating growth hormone receptor expression
11319536, Nov 06 2015 Ionis Pharmacueticals, Inc. Modulating apolipoprotein (a) expression
11332733, Feb 12 2018 lonis Pharmaceuticals, Inc. Modified compounds and uses thereof
11332746, Jun 27 2018 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing LRRK2 expression
11332748, Mar 18 2016 Caris Science, Inc. Oligonucleotide probes and uses thereof
11359197, Jan 12 2018 Bristol-Myers Squibb Company Antisense oligonucleotides targeting alpha-synuclein and uses thereof
11364304, Aug 25 2016 Northwestern University Crosslinked micellar spherical nucleic acids
11365416, Apr 11 2018 IONIS PHARMACEUTICALS, INC. Modulators of EZH2 expression
11376273, Oct 15 2008 IONIS PHARMACEUTICALS, INC. Modulation of factor 11 expression
11400161, Oct 06 2016 IONIS PHARMACEUTICALS, INC. Method of conjugating oligomeric compounds
11407997, Feb 22 2017 CRISPR THERAPEUTICS AG Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
11408000, Jun 03 2020 TAKEDA PHARMACEUTICALS U S A , INC Oligonucleotides for the treatment of nucleotide repeat expansion disorders associated with MSH3 activity
11433131, May 11 2017 Northwestern University Adoptive cell therapy using spherical nucleic acids (SNAs)
11434488, May 09 2018 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing ATXN3 expression
11447521, Nov 18 2020 Ionis Pharmaceuticals, Inc Compounds and methods for modulating angiotensinogen expression
11447775, Jan 12 2018 Bristol-Myers Squibb Company; Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting alpha-synuclein and uses thereof
11459564, Dec 21 2017 The Board of Regents of the University of Texas System Modulation of frataxin expression
11459587, Jul 06 2016 VERTEX PHARMACEUTICALS INCORPORATED Materials and methods for treatment of pain related disorders
11466279, Apr 09 2014 The Scripps Research Institute Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
11479530, Sep 24 2015 The Regents of the University of California; The Université de Montréal Synthetic sphingolipid-like molecules, drugs, methods of their synthesis and methods of treatment
11512143, Oct 30 2015 Genentech, Inc. Anti-HtrA1 antibodies and methods of use thereof
11530411, Jan 05 2016 IONIS PHARMACEUTICALS, INC. Methods for reducing LRRK2 expression
11535849, Apr 29 2010 IONIS PHARMACEUTICALS, INC. Modulation of transthyretin expression
11542504, May 01 2020 IONIS PHARMACEUTICALS, INC. Compounds and methods for modulating ATXN1
11547718, Nov 14 2018 Ionis Pharmaceuticals, Inc Modulators of FOXP3 expression
11559588, Feb 22 2017 CRISPR THERAPEUTICS AG Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
11564997, Jun 29 2016 CRISPR THERAPEUTICS AG Materials and methods for treatment of friedreich ataxia and other related disorders
11566245, Apr 20 2012 IONIS PHARMACEUTICALS, INC. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
11578372, Nov 05 2012 FOUNDATION MEDICINE, INC NTRK1 fusion molecules and uses thereof
11583548, Nov 10 2016 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing ATXN3 expression
11591595, Jul 19 2013 BIOGEN MA INC. Compositions for modulating Tau expression
11622993, Aug 03 2017 SYNTHORX, INC. Cytokine conjugates for the treatment of autoimmune diseases
11629348, Aug 15 2019 IONIS PHARMACEUTICALS, INC. Linkage modified oligomeric compounds and uses thereof
11633503, Jan 08 2009 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
11634451, Aug 08 2013 The Scripps Research Institute Method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
11634711, May 17 2012 IONIS PHARMACEUTICALS, INC. Methods and compositions for modulating apolipoprotein (a) expression
11661601, Mar 22 2018 IONIS PHARMACEUTICALS, INC.; The Regents of the University of Michigan Methods for modulating FMR1 expression
11690920, Jul 13 2017 Northwestern University General and direct method for preparing oligonucleotide-functionalized metal-organic framework nanoparticles
11701407, Aug 03 2017 SYNTHORX, INC. Cytokine conjugates for the treatment of proliferative and infectious diseases
11708614, Jun 15 2016 STRECK LLC Assays and methods for determining microbial resistance
11713462, Jun 17 2016 IONIS PHARMACEUTICALS, INC.; The Hospital for Sick Children Modulation of GYS1 expression
11725023, Jul 28 2015 Caris Science, Inc. Therapeutic oligonucleotides
11725208, Dec 14 2017 IONIS PHARMACEUTICALS, INC. Conjugated antisense compounds and their use
11732260, Mar 02 2018 IONIS PHARMACEUTICALS, INC.; Rosalind Franklin University of Medicine and Science Compounds and methods for the modulation of amyloid-β precursor protein
11732261, Aug 11 2011 IONIS PHARMACEUTICALS, INC. Selective antisense compounds and uses thereof
11732263, Jun 29 2020 Ionis Pharmaceuticals, Inc Compounds and methods for modulating PLP1
11732265, May 01 2014 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating complement factor B expression
11753460, Dec 13 2016 SEATTLE CHILDREN S HOSPITAL DBA SEATTLE CHILDREN S RESEARCH INSTITUTE Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
11753644, Jun 18 2021 Ionis Pharmaceuticals, Inc Compounds and methods for reducing IFNAR1 expression
11761007, Dec 18 2015 The Scripps Research Institute Production of unnatural nucleotides using a CRISPR/Cas9 system
11771698, Jan 18 2013 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
11781135, Mar 30 2012 Biogen MA Inc Methods for treating Alzheimer's disease
11786546, Jul 26 2019 Ionis Pharmaceuticals, Inc Compounds and methods for modulating GFAP
11801313, Jul 06 2016 VERTEX PHARMACEUTICALS INCORPORATED Materials and methods for treatment of pain related disorders
11833168, Jun 14 2018 IONIS PHARMACEUTICALS, INC.; Ludwig Institute for Cancer Research Compounds and methods for increasing STMN2 expression
11833221, Sep 01 2021 Ionis Pharmaceuticals, Inc Oligomeric compounds for reducing DMPK expression
11834479, Jun 24 2016 The Scripps Research Institute Nucleoside triphosphate transporter and uses thereof
11834660, Mar 19 2014 IONIS PHARMACEUTICALS, INC. Compositions for modulating Ataxin 2 expression
11834689, Jul 11 2017 The Scripps Research Institute; SYNTHORX, INC. Incorporation of unnatural nucleotides and methods thereof
11851653, Dec 01 2015 CRISPR THERAPEUTICS AG Materials and methods for treatment of alpha-1 antitrypsin deficiency
11851655, May 01 2013 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating apolipoprotein (a) expression
11859180, May 17 2012 IONIS PHARMACEUTICALS, INC. Antisense oligonucleotide compositions
11866727, Nov 06 2015 CRISPR THERAPEUTICS AG Materials and methods for treatment of glycogen storage disease type 1A
11873495, Jun 27 2018 IONIS PHARMACEUTICALS, INC. Compounds and methods for reducing LRRK2 expression
11879145, Jun 14 2019 The Scripps Research Institute Reagents and methods for replication, transcription, and translation in semi-synthetic organisms
11883535, Dec 03 2013 Northwestern University; Exicure, Inc. Liposomal particles, methods of making same and uses thereof
11884717, Nov 19 2015 The Brigham and Women's Hospital, Inc.; THE BROAD INSTITUTE, INC.; Massachusetts Institute of Technology Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
11898176, Apr 27 2012 Duke University Genetic correction of mutated genes
11905526, Oct 13 2015 Duke University Genome engineering with Type I CRISPR systems in eukaryotic cells
11912994, Apr 07 2015 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
5948903, May 08 1998 ISIS Pharmaceuticals, Inc. Synthesis of 3-deazapurines
6127124, Jan 20 1999 Isis Pharmaceuticals, Inc Fluorescence based nuclease assay
6147200, Aug 19 1999 Ionis Pharmaceuticals, Inc 2'-O-acetamido modified monomers and oligomers
6335434, Jun 16 1998 Isis Pharmaceuticals, Inc Nucleosidic and non-nucleosidic folate conjugates
6492111, Nov 25 1998 Isis Pharmaceuticals, Inc In situ binary synthesis of biologically effective molecules
6528631, Sep 03 1993 Isis Pharmaceuticals, Inc Oligonucleotide-folate conjugates
6656730, Jun 15 1999 Ionis Pharmaceuticals, Inc Oligonucleotides conjugated to protein-binding drugs
6680172, May 16 2000 REGENTS OF THE UNIVERSITY OF MICHIGAN, THE Treatments and markers for cancers of the central nervous system
6716625, Apr 16 1997 MYCOLOGICS, INC Histidine kinases of Aspergillus and other fungal species, related compositions, and methods of use
6762169, Jun 15 1999 Isis Pharmaceuticals, Inc Ligand-conjugated oligomeric compounds
6849723, Jun 06 1995 ISIS Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
7002006, Feb 12 2003 Ionis Pharmaceuticals, Inc Protection of nucleosides
7169916, Apr 01 2002 Ionis Pharmaceuticals, Inc Chloral-free DCA in oligonucleotide synthesis
7276599, Jun 02 2003 Ionis Pharmaceuticals, Inc Oligonucleotide synthesis with alternative solvents
7399845, Jan 27 2006 Ionis Pharmaceuticals, Inc 6-modified bicyclic nucleic acid analogs
7427675, Aug 23 2004 Ionis Pharmaceuticals, Inc Compounds and methods for the characterization of oligonucleotides
7507810, Feb 25 2005 Isis Pharmaceuticals, Inc Compositions and their uses directed to IL-4R alpha
7547684, May 11 2006 Ionis Pharmaceuticals, Inc 5′-modified bicyclic nucleic acid analogs
7666854, May 11 2006 Ionis Pharmaceuticals, Inc Bis-modified bicyclic nucleic acid analogs
7695902, Jun 06 1996 ISIS Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
7741457, Jan 27 2006 Ionis Pharmaceuticals, Inc 6-modified bicyclic nucleic acid analogs
7749504, Jun 20 2001 MS Technologies, LLC Anti-TAT188 antibodies
7750131, May 11 2006 Ionis Pharmaceuticals, Inc 5′-modified bicyclic nucleic acid analogs
7754450, Nov 15 2002 EISAI, INC Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
7759480, Apr 01 2002 Ionis Pharmaceuticals, Inc Chloral-free DCA in oligonucleotide synthesis
7786292, May 03 2006 Baltic Technology Development, Ltd. Antisense agents combining strongly bound base-modified oligonucleotide and artificial nuclease
7803915, Jun 20 2001 Genentech, Inc Antibody compositions for the diagnosis and treatment of tumor
7807372, Jun 04 2007 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
7812149, Jun 06 1996 Isis Pharmaceuticals, Inc 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
7875733, Sep 18 2003 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising 4′-thionucleosides for use in gene modulation
7884086, Sep 08 2004 Ionis Pharmaceuticals, Inc Conjugates for use in hepatocyte free uptake assays
7939677, Sep 18 2003 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising 4′-thionucleosides for use in gene modulation
7951785, Sep 21 2007 CHILDREN S HOSPITAL, LOS ANGELES; CHILDRENS HOSPITAL, LOS ANGELES; California Institute of Technology NFIA in glial fate determination, glioma therapy and astrocytoma treatment
8022046, Apr 18 2008 University of Pittsburgh - Of the Commonwealth System of Higher Education Microsphere-based composition for preventing and/or reversing new-onset autoimmune diabetes
8030467, May 11 2006 Ionis Pharmaceuticals, Inc 5′-modified bicyclic nucleic acid analogs
8088746, May 11 2006 Ionis Pharmaceuticals, Inc Bis-modified bicyclic nucleic acid analogs
8101743, Apr 05 2004 Ionis Pharmaceuticals, Inc Modulation of transthyretin expression
8129515, Jan 27 2006 Ionis Pharmaceuticals, Inc Oligomeric compounds and compositions for the use in modulation of microRNAs
8153603, Feb 25 2005 Isis Pharmaceuticals, Inc Compositions and their uses directed to IL-4R alpha
8188059, May 05 2006 Isis Pharmaceuticals, Inc Compounds and methods for modulating expression of GCGR
8198253, Jul 19 2006 Isis Pharmaceuticals, Inc Compositions and their uses directed to HBXIP
8252756, Jun 14 2005 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
8268980, May 11 2006 Ionis Pharmaceuticals, Inc 5′-modified bicyclic nucleic acid analogs
8278283, Jul 05 2007 Ionis Pharmaceuticals, Inc 6-disubstituted or unsaturated bicyclic nucleic acid analogs
8278426, Jun 08 2007 Ionis Pharmaceuticals, Inc Carbocyclic bicyclic nucleic acid analogs
8362232, May 05 2006 ISIS Pharmaceuticals, Inc. Compounds and methods for modulating expression of SGLT2
8372967, May 05 2006 Isis Pharmaceuticals, Inc Compounds and methods for modulating expression of GCCR
8394947, Mar 03 2004 Ionis Pharmaceuticals, Inc Positionally modified siRNA constructs
8426378, Mar 21 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising tricyclic nucelosides and methods for their use
8445229, Nov 15 2002 EISAI, INC Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
8501805, Sep 24 2008 Ionis Pharmaceuticals, Inc Substituted alpha-L-bicyclic nucleosides
8507200, Feb 09 2007 Northwestern University Particles for detecting intracellular targets
8530640, Feb 07 2008 Ionis Pharmaceuticals, Inc Bicyclic cyclohexitol nucleic acid analogs
8569474, Mar 08 2004 Ionis Pharmaceuticals, Inc Double stranded constructs comprising one or more short strands hybridized to a longer strand
8604183, Nov 05 2002 Ionis Pharmaceuticals, Inc Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
8604192, Sep 24 2008 Ionis Pharmaceuticals, Inc Cyclohexenyl nucleic acids analogs
8673871, May 05 2006 Ionis Pharmaceuticals, Inc Compounds and methods for modulating expression ApoB
8697860, Apr 29 2010 Ionis Pharmaceuticals, Inc Diagnosis and treatment of disease
8703728, Sep 09 2003 Ionis Pharmaceuticals, Inc Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
8779118, Jan 11 2010 Ionis Pharmaceuticals, Inc Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
8846637, Jun 08 2010 Ionis Pharmaceuticals, Inc Substituted 2′-amino and 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
8846639, Apr 04 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
8883752, Oct 24 2008 Ionis Pharmaceuticals, Inc 5′ and 2′ BIS-substituted nucleosides and oligomeric compounds prepared therefrom
8889350, Mar 26 2010 Integrated DNA Technologies, Inc Methods and compositions for isolating polynucleotides
8957200, Jun 07 2010 Ionis Pharmaceuticals, Inc Bicyclic nucleosides and oligomeric compounds prepared therefrom
8969316, May 05 2006 ISIS Pharmaceuticals, Inc. Compounds and methods for modulating expression of DGAT2
8987435, Oct 24 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds and methods
8993528, Mar 21 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising tricyclic nucelosides and methods for their use
8993738, Apr 28 2010 Ionis Pharmaceuticals, Inc Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
8999947, Jun 14 2005 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
9012421, Aug 06 2009 Ionis Pharmaceuticals, Inc Bicyclic cyclohexose nucleic acid analogs
9029335, Oct 16 2012 Ionis Pharmaceuticals, Inc Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
9045754, May 05 2006 Ionis Pharmaceuticals, Inc Short antisense compounds with gapmer configuration
9061044, Apr 29 2010 Ionis Pharmaceuticals, Inc Modulation of transthyretin expression
9096636, Jun 06 1996 Isis Pharmaceuticals, Inc Chimeric oligomeric compounds and their use in gene modulation
9127272, Jan 27 2006 Ionis Pharmaceuticals, Inc Oligomeric compounds and compositions for the use in modulation of target nucleic acids
9127276, May 01 2013 Ionis Pharmaceuticals, Inc Conjugated antisense compounds and their use
9139827, Nov 24 2008 Northwestern University Polyvalent RNA-nanoparticle compositions
9145558, May 01 2013 Glaxo Group Limited; Ionis Pharmaceuticals, Inc Compositions and methods for modulating HBV expression
9150606, Nov 05 2002 Ionis Pharmaceuticals, Inc Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
9156873, Apr 28 2010 Ionis Pharmaceuticals, Inc Modified 5′ diphosphate nucleosides and oligomeric compounds prepared therefrom
9163239, May 01 2013 Ionis Pharmaceuticals, Inc Compositions and methods for modulating apolipoprotein C-III expression
9181549, May 01 2013 Ionis Pharmaceuticals, Inc Conjugated antisense compounds and their use
9181550, May 01 2013 Ionis Pharmaceuticals, Inc Compositions and methods for modulating apolipoprotein (a) expression
9205102, Dec 06 2013 The University of British Columbia Method for treatment of castration-resistant prostate cancer
9221864, Apr 09 2012 Ionis Pharmaceuticals, Inc Tricyclic nucleic acid analogs
9243291, Dec 01 2011 Ionis Pharmaceuticals, Inc Methods of predicting toxicity
9290534, Apr 04 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds having at least one neutrally linked terminal bicyclic nucleoside
9321799, Apr 28 2010 Ionis Pharmaceuticals, Inc Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
9347095, Mar 15 2013 BIO-RAD LABORATORIES, INC Digital assays for mutation detection
9353371, May 02 2011 Ionis Pharmaceuticals, Inc Antisense compounds targeting genes associated with usher syndrome
9376690, Oct 30 2009 Northwestern University Templated nanoconjugates
9382540, May 01 2014 IONIS PHARMACUETICALS, INC Compositions and methods for modulating angiopoietin-like 3 expression
9394333, Dec 02 2008 WAVE LIFE SCIENCES LTD Method for the synthesis of phosphorus atom modified nucleic acids
9399774, Apr 29 2010 Ionis Pharmaceuticals, Inc Modulation of transthyretin expression
9403865, Aug 15 2012 Ionis Pharmaceuticals, Inc Method of preparing oligomeric compounds using modified capping protocols
9409934, Sep 24 2008 Ionis Pharmaceuticals, Inc Cyclohexenyl nucleic acids analogs
9428750, Mar 21 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising tricyclic nucleosides and methods for their use
9487780, Jun 01 2012 Ionis Pharmaceuticals, Inc Antisense compounds targeting genes associated with fibronectin
9499817, Sep 06 2012 The University of Chicago Antisense polynucleotides to induce exon skipping and methods of treating dystrophies
9506056, Jun 08 2006 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
9518259, Jun 15 2010 Ionis Pharmaceuticals, Inc Compounds and methods for modulating interaction between proteins and target nucleic acids
9556434, Oct 20 2010 Rosalind Franklin University of Medicine and Science Antisense oligonucleotides that target a cryptic splice site in Ush1c as a therapeutic for usher syndrome
9574193, May 17 2012 Ionis Pharmaceuticals, Inc Methods and compositions for modulating apolipoprotein (a) expression
9593374, Aug 17 2005 ONCOLAB DIAGNOSTICS GMBH Composition and method for determination of CK19 expression
9598458, Jul 13 2012 WAVE LIFE SCIENCES LTD Asymmetric auxiliary group
9598693, Jan 27 2006 Ionis Pharmaceuticals, Inc Oligomeric compounds and compositions for the use in modulation of micrornas
9605019, Jul 19 2011 WAVE LIFE SCIENCES LTD Methods for the synthesis of functionalized nucleic acids
9617541, Aug 20 2014 Northwestern University Biocompatible infinite coordination polymer nanoparticle-nucleic acid conjugates for antisense gene regulation
9617547, Jul 13 2012 WAVE LIFE SCIENCES JAPAN, INC Chiral nucleic acid adjuvant
9644207, Mar 14 2013 Ionis Pharmaceuticals, Inc Compositions and methods for modulating Tau expression
9677075, Apr 23 2013 Northwestern University Metal-ligand coordination polymer nanoparticles and methods for making
9683235, Jul 19 2013 Biogen MA Inc Compositions for modulating Tau expression
9688707, Dec 30 2014 Ionis Pharmaceuticals, Inc Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
9695211, Dec 02 2008 WAVE LIFE SCIENCES LTD Method for the synthesis of phosphorus atom modified nucleic acids
9695418, Oct 11 2012 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising bicyclic nucleosides and uses thereof
9695475, Dec 11 2012 Ionis Pharmaceuticals, Inc Competitive modulation of microRNAs
9714421, May 01 2013 Ionis Pharmaceuticals, Inc Compositions and methods
9719089, Jun 14 2005 Northwestern University Nucleic acid functionalized nonoparticles for therapeutic applications
9738727, Oct 14 2011 F HOFFMANN-LA ROCHE AG Anti-HtrA1 antibodies and methods of use
9738895, Oct 24 2008 Ionis Pharmaceuticals, Inc Oligomeric compounds and methods
9744183, Jul 06 2009 WAVE LIFE SCIENCES PTE LTD Nucleic acid prodrugs and methods of use thereof
9752142, Aug 11 2011 Ionis Pharmaceuticals, Inc Gapped oligomeric compounds comprising 5′-modified deoxyribonucleosides in the gap and uses thereof
9757475, Oct 30 2009 Northwestern University Templated nanoconjugates
9765338, Jul 19 2010 Ionis Pharmaceuticals, Inc Modulation of dystrophia myotonica-protein kinase (DMPK) expression
9777271, Sep 06 2012 The University of Chicago Antisense polynucleotides to induce exon skipping and methods of treating dystrophies
9803195, Dec 28 2005 University of Florida Board of Trustees; University of Florida Research Foundation Natural antisense and non-coding RNA transcripts as drug targets
9828602, Jun 01 2012 Ionis Pharmaceuticals, Inc Antisense compounds targeting genes associated with fibronectin
9844562, Nov 24 2008 Northwestern University Polyvalent RNA-nanoparticle compositions
9885037, Jul 13 2012 WAVE LIFE SCIENCES LTD Chiral control
9889209, Sep 14 2011 Northwestern University Nanoconjugates able to cross the blood-brain barrier
9890427, Feb 09 2007 Northwestern University Particles for detecting intracellular targets
9896709, Mar 13 2012 Integrated DNA Technologies, Inc Methods and compositions for size-controlled homopolymer tailing of substrate polynucleotides by a nucleic acid polymerase
9914922, Apr 20 2012 Ionis Pharmaceuticals, Inc Oligomeric compounds comprising bicyclic nucleotides and uses thereof
9926556, Apr 28 2014 IONIS PHARMACEUTICALS, INC. Linkage modified oligomeric compounds
9932580, May 01 2013 Glaxo Group Limited; Ionis Pharmaceuticals, Inc Compositions and methods for modulating HBV expression
9932581, May 01 2013 Ionis Pharmaceuticals, Inc Compositions and methods for modulating apolipoprotein C-III expression
9943604, Sep 20 2013 IONIS PHARMACEUTICALS, INC. Targeted therapeutic nucleosides and their use
9957292, May 01 2014 Ionis Pharmaceuticals, Inc Compositions and methods for modulating angiopoietin-like 3 expression
9957504, May 01 2013 Ionis Pharmaceuticals, Inc Compositions and methods for modulating apolipoprotein (a) expression
9976138, Aug 29 2011 Ionis Pharmaceuticals, Inc Methods and compounds useful in conditions related to repeat expansion
9982257, Jul 13 2012 WAVE LIFE SCIENCES LTD Chiral control
9994855, May 01 2014 IONIS PHARMACEUTICALS, INC. Compositions and methods for modulating growth hormone receptor expression
Patent Priority Assignee Title
3919193,
4381344, Apr 25 1980 Burroughs Wellcome Co. Process for producing deoxyribosides using bacterial phosphorylase
4481195, Oct 13 1978 CENTURY SCIENCE CORP , A CORP OF DE Method for the treatment of tumors with β-glucuronidase activity dependent pharmaceuticals
5457191, Jan 11 1990 Isis Pharmaceuticals, Inc 3-deazapurines
5587470, Jan 19 1990 ISIS Pharmaceuticals, Inc. 3-deazapurines
WO9110671,
////
Executed onAssignorAssigneeConveyanceFrameReelDoc
Jun 26 1995ACEVEDO, OSCAR L Isis Pharmaceuticals, IncCROSS-REFERENCING OF ASSIGNMENT FROM PRIOR APPLICATION S N 08 027,011 FILED 3 5 93, RECORDED AT REEL FRAME 7734 00060087630159 pdf
Jun 26 1995ANDREWS, ROBERT S Isis Pharmaceuticals, IncCROSS-REFERENCING OF ASSIGNMENT FROM PRIOR APPLICATION S N 08 027,011 FILED 3 5 93, RECORDED AT REEL FRAME 7734 00440087630171 pdf
Jun 26 1995COOK, PHILLIP DANIsis Pharmaceuticals, IncCROSS-REFERENCING OF ASSIGNMENT FROM PRIOR APPLICATION S N 08 027,011 FILED 3 5 93, RECORDED AT REEL FRAME 7734 0025 0087640108 pdf
Dec 10 1996ISIS Pharmaceuticals, Inc.(assignment on the face of the patent)
Date Maintenance Fee Events
Sep 28 2001M283: Payment of Maintenance Fee, 4th Yr, Small Entity.
Sep 27 2005M2552: Payment of Maintenance Fee, 8th Yr, Small Entity.
Dec 14 2009REM: Maintenance Fee Reminder Mailed.
May 12 2010EXP: Patent Expired for Failure to Pay Maintenance Fees.


Date Maintenance Schedule
May 12 20014 years fee payment window open
Nov 12 20016 months grace period start (w surcharge)
May 12 2002patent expiry (for year 4)
May 12 20042 years to revive unintentionally abandoned end. (for year 4)
May 12 20058 years fee payment window open
Nov 12 20056 months grace period start (w surcharge)
May 12 2006patent expiry (for year 8)
May 12 20082 years to revive unintentionally abandoned end. (for year 8)
May 12 200912 years fee payment window open
Nov 12 20096 months grace period start (w surcharge)
May 12 2010patent expiry (for year 12)
May 12 20122 years to revive unintentionally abandoned end. (for year 12)